≈ ≈

Transcription

≈ ≈
CONTENTS
TRANSACTIONS
AMERICAN BRONCHO-ESOPHAGOLOGICAL
ASSOCIATION
2010
≈
NINETIETH ANNUAL MEETING
PARIS/BALLY’S LAS VEGAS
LAS VEGAS, NEVADA
APRIL 28-29, 2010
≈
PUBLISHED BY THE ASSOCIATION
CONTENTS
Annual Photograph ..............................................................................................................................................................5
Officers 2009-2010 ..............................................................................................................................................................6
Executive Sessions
Minutes ........................................................................................................................................................................7
Reports .........................................................................................................................................................................7
Officers 2010-2011 ............................................................................................................................................................17
Presidential Address.
Andrew Blitzer, MD, DDS ........................................................................................................................................18
Presentation of Guest of Honor William Lawson, MD
Andrew Blitzer, MD, DDS ........................................................................................................................................19
Presentation of Chevalier Jackson Award to Clarence T. Sasaki, MD
Andrew Blitzer, MD, DDS ........................................................................................................................................20
Guests of Honor 1951-2010 ...............................................................................................................................................21
Recipients of Chevalier Jackson Award .............................................................................................................................21
Introduction of Chevalier Jackson Lecturer Marshall Strome, MD
Andrew Blitzer, MD, DDS ........................................................................................................................................22
Chevalier Jackson Lectures 1964-2010 .............................................................................................................................23
Presidential Citations Honoring Anthony F. Jahn, MD, Mitchell F. Brin, MD, and Robert M. Blitzer, DDS
Andrew Blitzer, MD, DDS ........................................................................................................................................24
President-Elect Address.
Michael A. Rothschild, MD .......................................................................................................................................27
SCIENTIFIC SESSIONS
ABEA Panel on Laryngopharyngeal Reflux
Moderator: Peter Belafsky, MD, PhD
Panelists: Jamie Koufman, MD; Tanya Meyer, MD; Nikki Johnston, PhD; Michael Pitman, MD ..........................28
Special Presidential Lecture
Airway Clinic: Bronchoesophagology in the Twenty-First Century
Marshall E. Smith, MD; Mark Elstad, MD................................................................................................................29
Arytenoid Abduction: Indications and Limitations
Gayle Woodson, MD..................................................................................................................................................31
Vibratory Asymmetry in Mobile Vocal Folds: Is It Predictive of Vocal Fold Paresis?
C. Blake Simpson, MD; Linda Seitan May, MD; Jill K. Green, MS;
Robert L. Eller, MD; Carlayne E. Jackson, MD ........................................................................................................38
Effects of Nerve-Muscle Pedicle on Immobile Rat Vocal Folds in the Presence of Partial Innervation
Takashi Aoyama, MD; Yoshihiko Kumai, MD, PhD; Eiji Yumoto, MD, PhD;
Takaaki Ito, MD, PhD; Satoru Miyamaru, MD, PhD ................................................................................................42
Use of a Microdebrider for Subepithelial Excision of Benign Vocal Fold Lesions
Alex Y. Cheng, MD; Ahmed M. S. Soliman, MD .....................................................................................................49
Response of Cricopharyngeus Muscle to Esophageal Stimulation by Mechanical Distension
and Acid and Bile Perfusion
Natalya Chernichenko, MD; Jeong-Soo Woo, MD; Jagdeep S. Hundal, MD; Clarence T. Sasaki, MD...................53
Analysis of Pepsin in Tracheoesophageal Puncture Sites
Jonathan M. Bock, MD; Mary K. Brawley, MA; Nikki Johnston, PhD; Tina Samuels, MS;
Becky L. Massey, MD; Bruce H. Campbell, MD; Robert J. Toohill, MD; Joel H. Blumin, MD..............................59
Low-Acid Diet for Recalcitrant Laryngopharyngeal Reflux: Therapeutic Benefits and Their Implications
Jamie A. Koufman, MD .............................................................................................................................................66
Cooling the “Oven”: A Temperature Study of Air and Glottal Tissue During Laser Surgery in an
Ex Vivo Calf Larynx Model
James A. Burns, MD; Gerardo Lopez-Guerra, MD; James T. Heaton, PhD; James B. Kobler, PhD;
Julie Kraas; Steven M. Zeitels, MD ...........................................................................................................................73
Quantity and Three-Dimensional Position of the Recurrent and Superior Laryngeal Nerve Lower
Motor Neurons in a Rat Model
Philip Weissbrod, MD; Michael J. Pitman, MD; Sansar Sharma, MD;
Aaron Bender; Steven D. Schaefer, MD ....................................................................................................................79
2
Contents
3
Assessment of Canine Vocal Fold Function After Injection of a New Biomaterial Designed to
Treat Phonatory Mucosal Scarring
Sandeep S. Karajanagi, PhD; Gerardo Lopez-Guerra, MD; Hyoungshin Park, PhD;
James B. Kobler, PhD; Marilyn Galindo; Jon Aanestad; Daryush D. Mehta, PhD;
Yoshihiko Kumai, MD, PhD; Nicholas Giordano; Anthony d’Almeida; James T. Heaton, PhD;
Robert Langer, ScD; Victoria L. M. Herrera, MD; William Faquin, MD, PhD;
Robert E. Hillman, PhD; Steven M. Zeitels, MD ......................................................................................................87
Potential of Induced Pluripotent Stem Cells for the Regeneration of the Tracheal Wall
Mitsuyoshi Imaizumi, MD; Yukio Nomoto, MD; Takashi Sugino, MD; Masao Miyake, PhD;
Ikuo Wada, PhD; Tatsuo Nakamura, MD; Koichi Omori, MD..................................................................................97
Determination of Predictive Factors of Tracheobronchial Prosthesis Removal: Stent Brands Are Crucial
Guillaume Buiret, MD; Carole Colin, MD; Guillaume Landry, MD;
Marc Poupart, MD; Jean-Christian Pignat, MD ......................................................................................................104
Posterior Glottic Diastasis: Mechanically Deceptive and Often Overlooked
Steven M. Zeitels, MD; Alessandro de Alarcon, MD; James A. Burns, MD;
Gerardo Lopez-Guerra, MD; Robert E. Hillman, PhD ............................................................................................111
Neurologic Variant Laryngomalacia Associated With Chiari Malformation and Cervicomedullary
Compression: Case Reports
Rajanya S. Petersson, MD; Nicholas M. Wetjen, MD; Dana M. Thompson, MD, MS...........................................121
Systematic Review of Endoscopic Airway Findings in Children With Gastroesophageal Reflux Disease
Jason Glenn May, MD; Priyanka Shah, MA; Lori Lemonnier, MD; Gaurav Bhatti, MS;
Jovana Koscica, DO; James M. Coticchia, MD.......................................................................................................126
Correlation of Vocal Fold Nodule Size in Children and Perceptual Assessment of Voice Quality
Roger C. Nuss, MD; Jessica Ward; Lin Huang, PhD; Mark Volk, DMD, MD;
Geralyn Harvey Woodnorth, MA.............................................................................................................................133
The Management of Postintubation Phonatory Insufficiency [Abstract]
Lindsey C. Arviso, MD; Adam M. Klein, MD; Michael M. Johns III, MD ............................................................138
Analysis of Vocal Fold Injection Amount: Association With Diagnosis and Outcome [Abstract]
VyVy N. Young, MD; Libby J. Smith, DO ..............................................................................................................138
Glidescope-Assisted Medialization Laryngoplasty: A Novel Technique [Abstract]
Roberta Lima, MD; Nazaneen Grant, MD...............................................................................................................139
Laryngeal Findings in Occluded Versus Unoccluded Ostia Model of Rhinosinusitis [Abstract]
Ankona Ghosh; Marcelo B. Antunes, MD; Joel Guss, MD; Fenella Long, PhD;
Noam A. Cohen, MD, PhD; Natasha Mirza, MD ....................................................................................................139
Narrowband Imaging and High-Definition Television for Detection, Definition, and Surveillance
of Head and Neck Cancer: A Prospective Study [Abstract]
Cesare Piazza, MD; Daniela Cocco, MD; Francesca Del Bon, MD; Stefano Mangili, MD;
Luigi de Benedetto, MD; Giorgio Peretti, MD ........................................................................................................140
Optimizing the Surgical Creation of a Supraglottal Sound Source: Theory and a Case Study [Abstract]
Sid Khosla, MD; Shanmugam Murugappan, PhD; Bernice Klaben, PhD...............................................................140
Primary TEP Placement in Patients With Laryngopharyngeal Free Tissue Reconstruction and Salivary
Bypass Tube Placement [Abstract]
Vasu Divi, MD; Derrick T. Lin, MD; Kevin Emerick, MD; Daniel G. Deschler, MD ............................................141
Development of the Dyspnea Severity Index–10 [Abstract]
Jackie Gartner-Schmidt, PhD, SLP; Adrianna Shembel, BS, MA; Priya Krishna, MD;
Rita Hersan, CCC-SLP; Thomas Zullo, PhD; Clark A. Rosen, MD .......................................................................141
Pepsin, at pH 7, in Nonacidic Laryngopharyngeal Refluxate, Significantly Alters the Expression of
Multiple Genes Implicated in Carcinogenesis [Abstract]
Nikki Johnston, PhD ................................................................................................................................................142
The Role of the Modified Barium Swallow Study and Esophagram in Patients With
GERD/Globus Sensation [Abstract]
Jayme R. Dowdall, MD; Tom Willis, MD; Adam J. Folbe, MD; James P. Dworkin, PhD .....................................142
Effect of Different Angiolytic Lasers Upon Resolution of Submucosal Hematoma in an
Animal Model [Abstract]
Daniel Novakovic, MD; Joanna D’Elia, MD; Andrew Blitzer, MD; Milton Waner, MD .......................................143
Characterization of Mesenchymal Stem Cells From Human Vocal Fold Fibroblasts [Abstract]
Summer E. Hanson, MD; Jaehyup Kim, MD; Beatriz H. Quinchia Johnson, DDS, PhD;
Melissa Breunig; Bridget Bradley; Peiman Hematti, MD; Susan L. Thibeault, PhD..............................................143
4
Contents
Utility of Diagnostic Testing in Adults With Idiopathic Subglottic Stenosis [Abstract]
Melissa McCarty Statham, MD; Yash A. Patil, MD; Allesandro de Alarcon, MD;
Ravindra G. Elluru, MD, PhD; Meredith E. Tabangin, MPH; Michael W. Bowen, PAC;
Thomas K. Chung, BS; Michael J. Rutter, MD, FRACS ........................................................................................144
A Challenging Case: Intubation Through a Foreign Body and Foreign Body Removal
From the Larynx [Abstract]
Vartan A. Mardirossian, MD; Timothy Anderson, MD; Joyce Colton-House, MD ................................................144
Calcium Oxalate Crystals Decode the Pulmonary Rhizopuzzle in a Pulmonary Abscess [Abstract]
Christopher G. Tang, MD; Christina C. Goette, MD; John Lin, MD;
Valerie Ng, MD, PhD; Alden H. Harken, MD .........................................................................................................145
In-Office Unsedated Transnasal Bronchoscopy for Removal of an Airway Foreign Body [Abstract]
Drew Plonk, MD; S. Carter Wright, Jr, MD; J. Whit Mims, MD; Catherine J. Rees, MD .....................................145
Endoscopic Management of a Penetrating Foreign Body in the Soft Tissue of the Retropharynx [Abstract]
Margaret L. Skinner, MD; Mark Volk, DMD, MD ..................................................................................................146
Delay in the Diagnosis of Laryngeal Cleft Associated With Tracheoesophageal Fistula [Abstract]
Margaret L. Skinner, MD; Stacey L. Ishman, MD; Umakanth Khatwa, MD;
Kenan Haver, MD; Rachel Rosen, MD; Craig Lillehei, MD; Reza Rahbar, MD....................................................146
Histopathologic Investigations of the Unphonated Human Child Vocal Fold Mucosa [Abstract]
Kiminori Sato, MD; Hirohito Umeno, MD; Tadashi Nakashima, MD;
Satoshi Nonaka, MD; Yasuaki Harabuchi, MD .......................................................................................................147
2010 Roster of Members
Honorary ..................................................................................................................................................................148
Senior .......................................................................................................................................................................148
Active .......................................................................................................................................................................149
Associate ..................................................................................................................................................................155
Corresponding ..........................................................................................................................................................155
Officers 1917-2010 ..........................................................................................................................................................157
MEMBERS IN ATTENDANCE AT THE NINETIETH ANNUAL MEETING
Row 1. Mark Courey, Clarence Sasaki, Gregory Grillone, Ellen Deutsch, Andrew Blitzer, Michael Rothschild, Gregory Postma,
Milan Amin, David Eibling, J. Scott McMurray.
Row 2. Michael Setzen, Gaelyn Garrett, Gayle Woodson, Stephen Conley, Ravindhra Elluru, Eiji Yanagisawa, Tadashi Nakashima,
Julie Wei, Charles Ford, Ellen Friedman, Gady Har-El, Jean Abitbol, Arnold Komisar, Marvin P. Fried.
Row 3. Edward Damrose, Natasha Mirza, Randal Paniello, Steven Zeitels, Mark Persky, Daniel Wohl, Alessandro de Alarcon,
Frederik G. Dikkers, Lauren Holinger, James Burns, Nick Maragos, Harvey Tucker, Stanley Shapshay.
Row 4. Craig Zalvan, Adam Klein, Paul Castellanos, Steffen Maune, Hirohito Umeno, Koichi Omori, Nicole Maronian, Shigeru
Hirano, Joel Blumin, Rob Stachler, Jim Cuyler, Libby Smith.
Row 5. Karen Zur, Ahmed Soliman, Thomas Murry, Lucian Sulica, Albert Merati, Kiminori Sato, Lee Akst, Steve Parnes, Peter
Koltai, Clark Rosen.
Row 6. Marshall Smith, Mario Andrea, Seth Dailey, Allan Abramson, Michael Johns.
5
OFFICERS 2009-2010
PRESIDENT
PRESIDENT-ELECT
ANDREW BLITZER, MD, DDS
New York, New York
MICHAEL A. ROTHSCHILD, MD
New York, New York
VICE-PRESIDENT
PAST PRESIDENT
ELLEN S. DEUTSCH, MD
Wilmington, Delaware
JAMIE A. KOUFMAN, MD
New York, New York
SECRETARY
TREASURER
GREGORY A. GRILLONE, MD
Boston, Massachusetts
GREGORY POSTMA, MD
Augusta, Georgia
EDITOR
CHAIR
AWARDS AND THESIS COMMITTEE
J. SCOTT MCMURRAY, MD
Madison, Wisconsin
CLARENCE T. SASAKI, MD
New Haven, Connecticut
CHAIR
NOMINATING COMMITTEE
CHAIR
FOREIGN BODY AND ACCIDENTS COMMITTEE
CLARENCE T. SASAKI, MD
New Haven, Connecticut
DANA THOMPSON, MD
Rochester, Minnesota
CHAIR
DIFFICULT AIRWAY COMMITTEE
CHAIR
PHARYNGEAL AND ESOPHAGEAL COMMITTEE
IAN N. JACOBS, MD
Philadelphia, Pennsylvania
MILAN AMIN, MD
New York, New York
CHAIR
INTERNATIONAL RELATIONS COMMITTEE
CHAIR
NEW TECHNOLOGY COMMITTEE
MARC J. REMACLE, MD, PHD
Yvoir, Belgium
J. SCOTT MCMURRAY, MD
Madison, Wisconsin
CHAIR
RESEARCH AND EDUCATION COMMITTEE
CHAIR
ONCOLOGY COMMITTEE
MARK S. COUREY, MD
Nashville, Tennessee
JAMES A. BURNS, MD
Boston, Massachusetts
REPRESENTATIVES TO THE BOARD OF GOVERNORS
DANA THOMPSON, MD
Rochester, Minnesota
GREGORY A. GRILLONE, MD
Boston, Massachusetts
CHAIR
SCIENTIFIC PROGRAM
JAMES A. BURNS, MD
Boston, Massachusetts
REPRESENTATIVE
AMERICAN ACADEMY OF OTOLARYNGOLOGY–
HEAD AND NECK SURGERY
COUNCIL MEMBERS AT LARGE
PETER BELAFSKY, MD
Sacramento, California
DAVID E. EIBLING, MD
Pittsburgh, Pennsylvania
IAN N. JACOBS, MD
Philadelphia, Pennsylvania
WEBMASTER
MICHAEL A. ROTHSCHILD, MD
New York, New York
6
Spring 2010 ABEA Council Meeting Minutes
Attending:
Milan Amin, MD
Peter Belafsky, MD
Andrew Blitzer, MD
James Burns, MD
Mark Courey, MD
Ellen Deutsch, MD
David Eibling, MD
Gregory Grillone, MD
Ian Jacobs, MD
Jamie Koufman, MD
Scott McMurray, MD
Gregory Postma, MD
Marc Remacle, MD
Michael Rothschild, MD
Clarence Sasaki, MD
The October 3, 2009, Council Meeting minutes were approved.
I. PRESIDENT’S REPORT — ANDREW BLITZER, MD
1. Hoarseness Guidelines
Discussion: The controversy that followed the publication of the guidelines has raised a number
of procedural and scientific issues in a public forum. Hopefully, other symptoms-based guidelines
that will be published in the future will not have the same kind of problems that occurred with
the hoarseness guidelines. This was in part a criticism of the guidelines on scientific grounds, but
more so a criticism of the process. Evidence-based medicine is important, but as someone at the
Harvard School of Public Health once said of otolaryngology, “...in your field there is no Level I
evidence to show that packing the nose stops nosebleeds because no one has done that study.” The
hope is that with the various responses from the ABEA and ALA there will be information that will
clearly neutralize and counterbalance the effect of what originally came out in the guidelines. The
hope is also that there will be enough good data out there to address any of the ill effects that may
have come from the Hoarseness Guidelines. In the interim we have probably changed the process
and established the ABEA in a greater way in laryngology. Dr Blitzer thanked all who have been
involved in this project.
Action Items:
a. Dr Eibling has been working on the rebuttal representing the ABEA. The document has gone
through several drafts and has been reviewed by Dr Postma (representing the ABEA), as well
as Dr Fried (representing the ALA) and Dr Blitzer (representing both the ABEA and the ALA).
There is a final document ready for Dr Rosenfeld, and we understand that he will publish it
as-is. Hopefully, this will end up in the “white journal” with both the ABEA and the ALA as
sponsors.
b. Dr Lucian Sulica has spearheaded the scientific response to the guidelines, which will be published in the Laryngoscope as a joint effort of the ABEA and ALA.
c. There will be a mini-seminar at the Academy meeting in Boston on “Hoarseness: Best Practices.” Hopefully, what comes out of that meeting will be published somewhere.
d. Dr Koufman acquired several web sites, including hoarsenessguidelines.com, which can be
used to post any rebuttal documents of position statements. When the ABEA comes up with
these documents, she would like permission to put it on-line.
2. ABEA Program
Discussion: Dr Blitzer thanked all the Council and said that the next 2 days will show what a strong
organization we have and what a great program we have put together.
Action Item: Each year, the outgoing President gives a gift to each Council member in appreciation of their support during their presidency. Dr Blitzer has decided to present a certificate of appreciation to each Council member and has made a $200 donation in the name of each Council
member toward the Past President’s Advisory Committee, headed by Dr Sasaki.
7
8
Minutes & Reports
3. ABEA/ALA/ELS Combined Meeting
Discussion: Dr Blitzer spoke with Dr Remacle about the combined meeting with the ABEA, ALA,
and ELS meeting at COSM. The tentative footprint would be that the ABEA and ALA would each
have a half-day session of their own and that on the second day both half-day sessions would be
devoted to the combined ABEA/ALA/ELS meeting. Another issue that needs to be resolved is the
registration process. The question is whether the ELS group could register once for both the combined meeting and the ABEA meeting. We would include ELS members on the Program Committee, panels, etc. COSM is in Chicago in 2011, in San Diego in 2012 (which would make for a much
longer flight for ELS members), and in Orlando in 2013. A final decision needs to be made about
which year we will hold the combined meeting. In return, the ABEA will encourage its members
to attend the ELS meetings.
Action Items:
a. Dr Grillone will be discussing this at the COSM Secretaries Liaison Committee meeting later
this week and will report back at the next ABEA Council meeting.
b. Dr Remacle will be discussing the combined meeting at the next ELS Executive Council and
will report back at the next ABEA Council meeting.
4. Committee Composition
Discussion: The ABEA is a very vibrant organization, and we have many members, including
younger members who would like to be more involved with the society. We need to make an effort to include them. To that end, Dr Grillone solicited the membership, asking for names of those
interested in becoming more involved. He distributed the list, as well as a list of current Committee Chairs and members. These lists were reviewed by the Council. The Council also reviewed the
progression table developed by Dr Deutsch to determine which committee chairs and members
had expired terms (see also President-Elect and Vice-President reports below).
Action Item: Dr Rothschild, as one of his first charges, will further review these documents and
draw from the list of interested members to appoint new committee chairs and other Council positions as appropriate.
5. New Committees
Discussion: In an effort to enrich the ABEA and expand representation within the Council, Dr
Blitzer has proposed three new committee changes that require approval by the Council. The new
committees are as follows.
Resident/Fellow Committee — for those residents and/or fellows who would like to play a more
active role in the ABEA. The chair would be a non-voting member of the Council. The charge
of the committee would be to bring to the Council issues pertinent to residents and fellows. This
would hopefully allow the infusion of new ideas from these younger members and plant the seed
for other residents to become interested in the ABEA. When they finish their training, they might
want to apply for active membership. The Academy has a Section for Residents and Fellows (SRF),
and they actually have one non-voting member on the Academy’s board of directors. There was
also discussion that resident membership should be a distinct category different from the category
of candidate member. The resident membership category would allow residents at any level to
become a member even before they were eligible for candidate membership. Any resident in good
standing could apply as a resident member with completion of an application without formal review by the Council. They would then go on the mailing list to receive the newsletter, and when
they completed residency they would be encouraged to become a candidate or active member. We
could communicate with the program chairs and ask them to advise any resident in an accredited
program that they could become a resident member by submitting a one-page application.
Past Presidents Advisory Committee — Many of the ABEA Past Presidents have spent many years
on the Council, and they still have much that they would like to and could offer to the society. This
would allow those Past Presidents who are interested in staying active in the society a chance to do
so. This would also help Dr Sasaki’s development efforts in the President’s Circle. The charge of
Minutes & Reports
9
this committee would be to provide senior-level advice to the Council. The chair of this committee would sit on the Council as an ex officio member. The chair will communicate with the other
Past Presidents prior to Council meetings and bring their comments to the meeting. They would
also have the option to meet separately as a group.
International Committee — When Dr Blitzer mentioned collaboration with the ELS in the last
Newsletter, Dr Yanagisawa wrote him to say that when he was President they met with the Japan
Laryngological Society in a collaborative effort. The International Liaison has been an integral part
of the Council, but this single position allows representation from only one region of the globe. An
International Committee would allow representation from other regions, such as South America,
Central America, and Asia. At the Academy, outside of Canada, Brazil has the largest number of
attendees. They have over 6,000 otolaryngologists, and they have almost 400 laryngologists. It
would be very advantageous to reach out and have representation from countries such as Brazil
on the International Committee. We encouraged the corresponding members several years ago to
become active. However, the By-Laws are not clear on the requirements for active membership of
international physicians. There is no distinction in the ELS, and their members just have to have
the appropriate board certification. The corresponding (ie, international) membership category was
originally established because of the difficulty that international members often had in traveling
long distances to attend meetings in the United States. That is no longer an issue, and many “corresponding” members come to many of our meetings. There should be one representative from
Asia, one from Europe, and one from South America on the committee. Dr Remacle, currently the
International Liaison, would serve as the first chair of the International Committee. The charge of
the committee would be to represent the interests of our international members.
Action Items:
a. Dr Grillone will look into clarifying the current requirements for candidate membership and
determine whether additional By-Laws language is necessary for the resident membership.
b. Dr Grillone and Dr Deutsch will to look into coming up with appropriate By-Laws language so
that the requirements for active membership are clarified and modified appropriately, allowing
international physicians to become active members.
Votes:
a. The council unanimously voted to 1) approve the formation of the Past President’s Advisory
Committee as an ad hoc committee and 2) to appoint Dr Blitzer (immediate Past President)
as the first chair of the committee.
b. The council unanimously voted to approve the formation of the Residents and Fellows Committee as an ad hoc committee. The incoming president will appoint the chair of the Residents Committee.
6. ABOto Certification Exams
Discussion: Dr Blitzer received a letter from Bob Miller, Executive Director of the ABOto, saying
that they have recently altered the blueprints of the ABOto exam to more accurately reflect individual otolaryngology practices. One component of each exam would be a clinical fundamentals
section that would test general knowledge that all otolaryngologists should have, regardless of
primary focus. Examples include airway management, tracheotomy, CPR, etc. Dr Miller asked the
ABEA to 1) comment on the list of topics as to their applicability to our specialty and 2) identify
individuals who would be interested in writing this clinical fundamentals curriculum and possibly
writing test questions. Drs Burns, Grillone, and McMurray indicated that they would be interested.
Action Item: Dr Blitzer will submit Drs Burns, Grillone, and McMurray’s names to Dr Miller.
7. Senior Membership Status for Pat Bradley
Discussion: Dr Pat Bradley has been a member since 1991 and wrote to Dr Blitzer to ask that he
be given emeritus status, as he is fully retired from practice and can’t afford dues but would still
10
Minutes & Reports
like to stay involved. Our senior member category requires that they have 20 years of membership
or be age 65 and request in writing that they be placed in senior status.
Vote: The Council unanimously voted to approve senior membership status for Dr Bradley.
8. Contract With the Annals
Discussion: Dr Blitzer said that we need to look at our relationship with the Annals and determine
whether we should continue that relationship. The ALA recently signed a new contract with the
Laryngoscope, making it the official journal of the ALA, so that all papers from the ALA meeting will be published in the Laryngoscope. One of the proposals that came out of the ALA was
to make either a new journal or a sub-journal, for example, a sub-journal of the Laryngoscope
just on laryngology or “airway issues,” which could include bronchology, laryngology, and upper airway disorders. Creating a separate journal from scratch runs the risk that authors may not
want to contribute because the impact is low. Dr Courey is on the ALA and ABEA councils and
could represent the ABEA with both journals (Annals and Laryngoscope) regarding this issue. Dr
McMurray said that the ABEA contract with the Annals started in 1994 and is renewed every 5
years. The current contract ends in 2014. The contract includes subscriptions to the Annals for the
members. The Annals publisher, Ken Cooper, informs us that this represents 252 print subscriptions and on-line access for the past 12 years (which he quoted out as a $240 value). He said that
was worth $125,000 in addition to the $7,000 a year with which we can do whatever we want. The
Annals publishes the Transactions at cost and mails them to the members. The Annals also gives
us free advertisement and free publicity, and they give us priority for publication of papers that
have been presented. Dr McMurray asked Ken Cooper about publishing a synopsis of the panel
discussions, and Ken thought that was a great idea. Dr McMurray also asked them for $2,500 to
support a panel, and Ken Cooper said in these economic times they didn’t feel they could do that.
Ken Cooper later sent Dr McMurray an e-mail and said that the Annals is the last independently
owned journal and that down the line the ABEA may want to have their own journal and perhaps
that would be a perfect fit. The amount of money they are giving us is less than what other journals
are giving. Dr Rothschild was at the board meeting of the Archives last month and reported that
it seems fairly clear that print publishing will go away in the not-too-distant future. Production is
expensive, and there are significant environmental impacts of printing and shipping.
Action Items:
a. Dr McMurray will speak with the Annals (Ken Cooper) to find out whether they would be willing to provide free journal subscriptions to the new class of resident members.
b. Dr Courey will investigate potential options with both publishers and report back after his meetings.
II. PRESIDENT-ELECT’S REPORT — MICHAEL ROTHSCHILD, MD
1. Committee Composition
Discussion: Dr Rothschild has worked with Drs Blitzer and Deutsch on the progression of the
Executive Council members. In accordance with the By-Laws, committee membership is for 3
years with the option for an additional 3-year term for committee chairs. Dr Deutsch prepared a
proposed progression table that shows that many of the chairs have been on for several years more
than the specified terms. If people are in their second term, they will be asked whether they want
to serve out their second term.
Dr Deutsch said that the time limit only went into effect when the By-Laws were revised 2 years
ago. She suggested that rather than make a lot of changes at once, we stagger those who have been
in their position the longest and perhaps have three new chairs a year. Proposed changes to the
standing committees are as follows.
a. Research and Education — Dr Courey has been chair for 7 years, and he will step down. Drs
Aviv and Grillone have been committee members. Dr Aviv has been President, and Dr Grillone
is currently Secretary. Dr Rothschild will select both the chair spot and the committee members.
Minutes & Reports
11
b. Difficult Airway — Dr Jacobs has been chair for 8 years and will step down. Dr Karen Zur will
become chairman.
c. New Technology — Dr McMurray has been chair for 9 years and will step down. Committee
members are Drs Dana Thompson, Michael Rutter, Paul Willging, and Andy Inglis. Dr Rothschild will ask one of these committee members whether they would like to be chair.
d. Foreign Body — Dr Thompson has been chair for 5 years and will stay if she wants. If not, Dr
Gresham Richter would be a good choice.
e. Oncology — Dr Burns has been chair for 7 years. He will stay.
f. International Liaison — Dr Remacle has been in this position for 6 years. He will become chair
of the newly proposed International Committee and will select other International Committee
members.
g. Pharyngoesophageal — Dr Amin has been chair for 3 years with no committee members. He
would like to stay on and select some committee members.
h. BOG Gov — Dr Grillone has been on for 6 years.
i. BOG PR — Dr McMurray has been on for 9 years and will step down.
j. BOG Legislative — Dr Thompson is in her second year and will stay.
k. Council at Large — Drs Belafsky and Eibling are the current Councilors-at-Large. Their terms
will expire in 2013.
Action Item: Dr Rothschild will further review the progression table and draw from the list of
interested members compiled by Dr Grillone to appoint new committee chairs and other Council
positions as appropriate.
2. Online Voting/Nominations for Officer Positions
Discussion: There was a discussion about the possibility of getting the slate of officers from the
Nominating Committee ahead of the Annual Meeting and then allowing members to vote on the
web site. We could also accept nominations from the membership prior to the announcement of the
selections from the Nominating Committee. This would allow members to vote if they are unable
to attend the Business Meeting. The President-Elect nominee should be selected from individuals
holding the position of Secretary, Treasurer, Editor, or Vice-President; this would be the pathway
to President. Committee chairs and Councilors-at-Large may move into one of the four officer
positions: Secretary, Treasurer, Editor, or Vice-President. It would be best to continue this policy,
as it assures continuity and institutional memory. Nominations from the membership would be for
the committee chairs and Councilors-at-Large. The Nominating Committee for this year will be Dr
Koufman as Past President, and the remainder of the Nominating Committee would be chosen at
the first Business Meeting. Dr Deutsch reported from the By-Laws that “This will consist of four
members, three of whom shall be nominated and elected the preceding year (we have been out of
compliance with this requirement) by the voting members at the executive session of ABEA and
the fourth member will be the immediate Past President.” We have accepted nominations at the
Business Meeting, and then they meet to present the slate the following day. Dr Koufman will select three members for the Nominating Committee this year. Dr Gregory Postma will be nominated
as President-Elect, and Dr Dana Thompson as Treasurer. Once we achieve compliance with the
By-Laws, there will be an opportunity to bring in one new person on the Council each year. Peter
Koltai was on the Council from 2001 to 2002 and served as Secretary for 4 years before moving
off. The Council felt that we should consider folding him back into succession for President. Dr
Koltai left the Council because of his commitment to serve as ASPO president, but he would like
to return to the Council once his term as ASPO president is up.
Action Item: The discussion regarding on-line voting was tabled and will be discussed further at
a later date.
3. Chevalier Jackson Stamp Project
Discussion: There was a discussion about the possibility of having a Chevalier Jackson postage
12
Minutes & Reports
stamp. Dr Wayne Hellman provided us with a historical essay published in the Newsletter last
year. The Post Office publishes stamps to honor famous Americans, and Dr Jackson would be
a good choice. He not only practiced bronchoscopy, but advanced the field of public health and
safety. Furthermore, he is a real American hero, because in 1916 the Europeans were still the predominant names in medicine, and he pushed back the frontiers of medicine from this side of the
Atlantic. Dr Hellman would be willing to help with this project and has a lot of knowledge on the
history of Dr Jackson.
III. VICE-PRESIDENT’S REPORT — ELLEN DEUTSCH, MD
1. Progression Table
Dr Deutsch’s progression table was discussed previously (see President-Elect’s report).
2. AAO-HNS Guidelines
Clinical Practice Guidelines continue to evolve. Some suggestions have been specialty-specific
guidelines. Dr Deutsch would be willing to help anyone who would like to submit a topic for consideration. Tonsillectomy may be the next Guideline.
3. Surgical Specialties Advocacy Meeting
There will be a surgical specialty advocacy meeting in Washington, DC, in July. Dr Jacobs volunteered to attend on behalf of the ABEA.
Action Item: Dr Jacobs will report back at the fall Council meeting.
IV. TREASURER’S REPORT — GREGORY POSTMA, MD
1. Financial Report
Dr Postma provided a financial report. We decided to let our investments ride through the recession. We had an increase of $1,150.32 since December 2008 in Wachovia Money Market, and an
increase of $25,105.72 in the Mellon Funds during the same period.
2. The President’s Circle
The President’s Circle was $15,000 as of December 31, 2009 but we are close to $25,000 now,
which is very encouraging.
V. EDITOR’S REPORT — SCOTT MCMURRAY, MD
Transactions
Discussion: Dr McMurray said the 2008 Transactions have been published and we are nearly ready
for the 2009 edition; all the papers are in. There are some additions he would like to add to the 2009
edition, and he will be sending some e-mails out. There was a discussion about publishing the Transactions on the web site, which would save publication and postage costs. All papers for 2010 should
have been submitted to Dr McMurray. If anyone has pictures for their presentations, please submit
them to Dr McMurray.
VI. SECRETARY’S REPORT — GREGORY GRILLONE, MD
1. SLC Update
Discussion: Dr Grillone reported that Dr Jerry Goldstein is stepping down this year as chairman
of the Secretaries Liaison Committee and that Dr Stanley Shapshay will be taking over. He will
attend the meeting this year and will take over officially after the meeting.
2. COSM Convention Cancellation Insurance
Discussion: Dr Grillone distributed a COSM convention cancellation insurance memo. This is
something the ACS has done in the past with the societies, and they will ask about it at the SLC
meeting this week. This is necessary to ensure against any forced cancellation or forced reduction
in attendance at meetings.
Vote: The Council voted unanimously to approve ABEA participation in the insurance plan.
Minutes & Reports
13
3. Future Meetings
Discussion: Dr Grillone discussed the schedule for the Annual Meeting for the coming 3 years.
The footprint is as follows.
a. 2011 — April 27–May 1 in Chicago.
b. 2012 — April 18-22 in San Diego.
c. 2013 — April 10-14 in Orlando.
4. Candidates for Membership
The following candidates were presented for membership and applications reviewed.
a. Fredrik Dikkers, MD.
b. Nanzeen Grant, MD.
c. Priya Krishna, MD.
d. Jeffrey Simons, MD.
e. Jean Verheyden.
f. Nwanmegha Young, MD.
g. Jonathan Bock, MD, has only been in practice for 2 years, so will be advised that he will be in
the candidate category this year and active next year.
VII. DEVELOPMENT OFFICER’S REPORT — CLARENCE SASAKI, MD
The President’s Circle
Discussion: Dr Sasaki prepared a letter to the membership outlining the purpose and success of the
President’s Circle, which will promote the science necessary in the care of our patients. As we go into
phase II of this development project, he suggested that the Council develop a succession of advisors.
He would like to continue for another year and see how it goes. There was a discussion about whether
these funds had been earmarked for any particular activity. Dr Sasaki said the ABEA has never had a
lot of money, and his original thought was that it would be used to support the general fund. We don’t
want to shackle these funds so we can’t use them, but we don’t want to make it too easy. Dr Sasaki
said that phase III could be to determine whether we need to establish an endowment.
VIII. PROGRAM COMMITTEE — JAMES BURNS, MD
1. Dr Burns thanked Drs Belafsky and Postma for moderating the panels.
2. The Council thanked Drs Burns and Blitzer for the program they have set up.
3. The panels will be recorded. They will give Leora a tape, which she will see that Dr Rothschild
will get.
IX. FOREIGN BODY COMMITTEE — DANA THOMPSON, MD
Dr Amin reported for Dr Thompson that there were four very good submissions and that they chose
one by Vartan A. Mardirossian, MD, of Boston, on “A Challenging Case: Intubation Through a Foreign Body and Foreign Body Removal From the Larynx.”
X. AWARDS AND THESIS COMMITTEE — CLARENCE SASAKI, MD
1. Dr Sasaki reported that the Broyles-Maloney Award had been given to Sandeep Karajanagi, PhD,
of Boston, for “Assessment of Canine Vocal Fold Function After Injection of a New Biomaterial
Designed to Treat Phonatory Mucosal Scarring.”
2. There were no Steven Gray or Seymour Cohen awards this year.
XI. PHARYNGEAL & ESOPHAGEAL COMMITTEE — MILAN AMIN, MD
Nothing to report.
14
Minutes & Reports
XII. NEW TECHNOLOGY — SCOTT MCMURRAY, MD
Dr McMurray thanked the Council for the 9 years he has served with them.
XIII. RESEARCH AND EDUCATION COMMITTEE — MARK COUREY, MD
Nothing to report.
XIV. INTERNATIONAL COMMITTEE — MARC REMACLE, MD
ABEA/ALA/ELS combined meeting (see President’s Report).
XV. BOARD OF GOVERNORS — GREGORY GRILLONE, MD
Reimbursement for Endoscopic Treatment of Zenker’s Diverticulum
Discussion: Dr Grillone discussed that he had been investigating reimbursement for this procedure.
In Massachusetts, many payers do not reimburse for this procedure, even though it is faster and requires less recovery time than open procedures. He discussed the problem with representatives from
the BOG, including Drs Schreibstein and Waguespack. The BOG indicated it might be a good idea if
the ABEA came up with a position statement for endoscopic Zenker’s diverticula.
Action Item: Drs Amin, Koufman, and Belafsky will work on the position statement and report at the
fall meeting.
XVI. ACS ADVISORY COMMITTEE — GREGORY GRILLONE, MD
Nothing to report.
XVII. WEBMASTER — MICHAEL ROTHSCHILD, MD
1. Dr Rothschild reported that he is able to host and maintain the ABEA web site. The site was recently overhauled so it is now Smartphone and iPhone accessible.
2. We are now sending the Newsletter out to the membership electronically, and it will be available
on-line in an electronic format. Back issues will be available as PDFs, as well.
3. We have the 2005 and 2006 Foreign Body Award clips on line. We also have the audio from the
2008 and 2009 COSM. Dr Rothschild encouraged anyone who has a video clip to submit it so he
can review it for placement on the site.
4. He has added Dr Sasaki’s fundraising letter, along with a downloadable form that can be used for
contributing to the ABEA.
XVIII. DIFFICULT AIRWAY COMMITTEE — IAN JACOBS, MD
1. Dr Jacobs said that they had their annual course with Dr Deutsch and Dr Zur, which went well.
They had registrants from the Northeast, and they are looking to expand it nationally. Dr Thompson is going to do it at Mayo, and Dr Gresham Richter is interested in doing it in South Carolina.
They are working on the IRBs.
2. The specialty committee (SSAC) will meet on Friday.
XIX. ONCOLOGY COMMITTEE — JAMES BURNS, MD
Dr Burns reported that the Oncology Committee was accepted to present an ABEA-sponsored miniseminar at the Academy. “State-of-the-Art Endoscopic Management of Larynx and Pharynx Cancer.”
XX. COUNCILORS-AT LARGE — PETER BELAFSKY, MD, AND DAVID EIBLING, MD
Voice-Lift Procedure
Discussion: Dr Belafsky said that in ENT Today there is a reference to treatment of dysphonia as a
cosmetic procedure. If we are going to have a position statement, it should emphasize the disability
associated with voice disorders in the elderly. Dr Koufman said she addressed this when the term
“voice lift” was originally used and that it should be abandoned. This is not a cosmetic procedure.
Dr Blitzer said he was contacted at the time by a reporter from the London Times who asked him to
comment on this procedure. He told them this was not a new concept. Augmentation of the vocal cord
Minutes & Reports
15
has been going on for many years. The only difference is that they have some new materials, but in
no way should it be suggested that when you are 60 years old you are going to have an operation and
sound like you did when you were 16. There are things we can do to improve voice, but such complete rejuvenation of the voice is an unrealistic expectation. Dr Koufman wanted the record to show
that she introduced bilateral medialization laryngoplasty in 1989. Before that, they weren’t doing
anything bilateral except catastrophic Teflon injections.
XXI. SURGICAL SUBSPECIALTIES ADVISORY COMMITTEE (SSAC) — IAN N. JACOBS, MD
Nothing to report.
XXII. NEW BUSINESS
Leora Loy was excused from the meeting while a discussion was held regarding her stipend. The
Council unanimously felt that she continues to do an outstanding job as liaison to the Secretary of the
society. In recognition of her efforts, the Council voted to approve a $1,500 bonus this year.
The Council meeting was adjourned at 6:30 PM.
ACCOUNTANTS’ COMPILATION REPORT
To the Board of Directors of the American
Broncho-Esophagological Association, Inc:
AMERICAN BRONCHO-ESOPHAGOLOGICAL
ASSOCIATION, INC
STATEMENT OF ASSETS AND UNRESTRICTED
NET ASSETS
CASH BASIS DECEMBER 31, 2009
We have compiled the accompanying statement
of assets and unrestricted net assets-cash basis of
the American Broncho-Esophagological Association, Inc (a nonprofit organization) as of December
31, 2009, and the related statement of unrestricted
revenues, expenses, and other changes in unrestricted net assets-cash basis for the year then ended, in
accordance with Statements on Standards for Accounting and Review Services issued by the American Institute of Certified Public Accountants. These
financial statements have been prepared on the cash
basis of accounting, which is a comprehensive basis of accounting other than generally accepted accounting principles.
ASSETS
Checking account
Savings account
President’s Circle account
Investments — Mellon Private Capital
Management
Total assets
TOTAL UNRESTRICTED NET ASSETS
$32,908
247,506
15,250
171,181
466,845
$466,845
See Accountants’ Compilation Report.
AMERICAN BRONCHO-ESOPHAGOLOGICAL
ASSOCIATION, INC
STATEMENT OF UNRESTRICTED REVENUES,
EXPENSES, AND OTHER CHANGES IN
UNRESTRICTED NET ASSETS
CASH BASIS FOR THE YEAR ENDED
DECEMBER 31, 2009
A compilation is limited to presenting in the form
of financial statements information that is the representation of management. We have not audited
or reviewed the accompanying financial statements
and, accordingly, do not express an opinion or any
other form of assurance on them. However, we did
become aware of a departure from the cash basis of
accounting that is described in the following paragraph.
REVENUES
Dues
Registration
Annals
Sponsorships and donations
Interest income
Investment income — Mellon Bank
Unrealized gain on investments — Mellon Bank
Total revenues
EXPENSES
Meetings
AAO-HNS council meeting
COSM meeting
Awards and grants
Chevalier Jackson
Broyles Maloney
Seymour Cohen
Secretarial support
Professional services
Bank service charges
Trustee fees
Foreign tax
Office supplies
Realized loss on sale of securities
Total expenses
CHANGE IN UNRESTRICTED NET ASSETS
UNRESTRICTED NET ASSETS
JANUARY 1, 2009
UNRESTRICTED NET ASSETS
DECEMBER 31, 2009
Investments have been stated at market value,
which is not considered a generally accepted modification of the cash basis of accounting. The effects
of this departure from the cash basis of accounting
on the accompanying financial statements have not
been determined.
Management has elected to omit substantially all
of the disclosures ordinarily included in financial
statements prepared on the cash basis of accounting. If the omitted disclosures were included in the
financial statements, they might influence the user’s
conclusions about the Organization’s financial position and changes in net assets. Accordingly, these
financial statements are not designed for those who
are not informed about such matters.
Piccerelli, Gilstein & Company, LLC
Providence, Rhode Island
June 21, 2010
See Accountants’ Compilation Report.
16
$37,565
16,315
7,000
15,250
1,150
3,408
28,213
108,901
2,012
15,896
2,500
1,500
1,000
9,500
4,369
248
489
20
119
6,029
43,682
65,219
401,626
$466,845
OFFICERS 2010-2011
PRESIDENT
PRESIDENT-ELECT
MICHAEL A. ROTHSCHILD, MD
New York, New York
GREGORY POSTMA, MD
Augusta, Georgia
VICE-PRESIDENT
PAST PRESIDENT
ELLEN S. DEUTSCH, MD
Wilmington, Delaware
ANDREW BLITZER, MD, DDS
New York, New York
SECRETARY
TREASURER
GREGORY A. GRILLONE, MD
Boston, Massachusetts
DANA THOMPSON, MD
Rochester, Minnesota
EDITOR
CHAIR
AWARDS AND THESIS COMMITTEE
J. SCOTT MCMURRAY, MD
Madison, Wisconsin
ANDREW BLITZER, MD, DDS
New York, New York
CHAIR
NOMINATING COMMITTEE
CHAIR
FOREIGN BODY AND ACCIDENTS COMMITTEE
JAMIE A. KOUFMAN, MD
New York, New York
GRESHAM RICHTER, MD
Little Rock, Arkansas
CHAIR
DIFFICULT AIRWAY COMMITTEE
CHAIR
PHARYNGEAL AND ESOPHAGEAL COMMITTEE
KAREN B. ZUR, MD
Philadelphia, Pennsylvania
MILAN AMIN, MD
New York, New York
CHAIR
INTERNATIONAL RELATIONS COMMITTEE
CHAIR
NEW TECHNOLOGY COMMITTEE
MARC J. REMACLE, MD, PHD
Yvoir, Belgium
PAUL WILLGING, MD
Cincinnati, Ohio
CHAIR
RESEARCH AND EDUCATION COMMITTEE
CHAIR
ONCOLOGY COMMITTEE
SETH DAILEY, MD
Madison, Wisconsin
JAMES A. BURNS, MD
Boston, Massachusetts
REPRESENTATIVES TO THE BOARD OF GOVERNORS
GREGORY A. GRILLONE, MD
Boston, Massachusetts
J. SCOTT MCMURRAY, MD
Madison, Wisconsin
DANA THOMPSON, MD
Rochester, Minnesota
CHAIR
SCIENTIFIC PROGRAM
KAREN B. ZUR, MD
Philadelphia, Pennsylvania
COUNCIL MEMBERS AT LARGE
REPRESENTATIVE
AMERICAN ACADEMY OF OTOLARYNGOLOGY–
HEAD AND NECK SURGERY
PETER BELAFSKY, MD
Sacramento, California
DAVID E. EIBLING, MD
Pittsburgh, Pennsylvania
REZA RAHBAR, MD
Boston, Massachusetts
IAN N. JACOBS, MD
Philadelphia, Pennsylvania
WEBMASTER
MICHAEL A. ROTHSCHILD, MD
New York, New York
17
PRESIDENTIAL ADDRESS
ANDREW BLITZER, MD, DDS
I welcome you to the 90th Annual Meeting of the
American Broncho-Esophagological Association, a
society that was started by Chevalier Jackson. I wish
to thank Dr James Burns, who worked tirelessly as
Program Chairman; Leora Loy, who serves as Administrator of the ABEA; and all of the council
members whose wisdom and guidance serve the society well.
In the Hippocratic oath that we all have taken
upon graduation from medical school, it says “...to
respect the hard-won scientific gains of those physicians in whose steps I walk and gladly share such
knowledge as is mine with those who follow.”
Jackson says in his autobiography, “Achievement
brings pleasure, but also the onerous feelings of
obligation; never refuse to treat patients in need of
care; the work of physicians in prevention of disease
should equal their work in curing disease; always
share and teach the skills you have learned to your
students; and always look for ways of giving back
for the honor of being a physician.”
Some of the ways that all of you can give back
are:
–Volunteer for the committees and help make the
organization better.
–Join the ABEA and contribute to the meetings.
–Donate money to the President’s Circle and
Foundation Fund to help support the meetings,
awards, and resident and fellow participation.
–Present your work at our meetings and publish
in our journal (the Annals).
18
PRESENTATION OF GUEST OF HONOR WILLIAM LAWSON, MD
ANDREW BLITZER, MD, DDS
William Lawson grew up in New York City and
received his DDS and MD degrees from New York
University. He did his oral surgery and otolaryngology training at the Mt Sinai Medical Center, with a
brief interruption as a Captain in the US Army Medical Corps. He is a Professor of Otolaryngology, and
has remained with the faculty of Mt Sinai Medical
Center for 36 years, training me and many of the
otolaryngologists in the audience. The breadth of his
knowledge is encyclopedic. His surgical skills and
teaching span much of our specialty, including head
and neck cancer, trauma and reconstructive surgery,
sinus and airway surgery, and facial plastic surgery.
He is the director of a facial plastic surgery fellowship, director of a sinus and airway disease laboratory, and member of the multi-discipline Head and
Neck Tumor Board. He is a fellow of 12 societies,
including the ABEA, American Laryngological Association, Triological Society, Society of Maxillofacial Surgery, and American Rhinologic Society. He
has received numerous honors and awards, including a Distinguished Candidate’s Thesis and Presidential Citation from the Triological Society. He has
held many offices in these societies, including on
the boards of directors of the American Academy of
Facial Plastic and Reconstructive Surgery and the
American Board of Facial Plastic Surgery, and as
President of the New York Laryngological Society.
He has taught courses and given lectures worldwide
on the subjects of head and neck disease and surgery, airway evaluation and surgery, and facial plastic surgery. His basic research is comprehensive and
spans results from his animal laboratory for sinus
and airway disease, the comparative and paleo anthropology of airway development (for which he is a
consultant to the Museum of Natural History in New
York), and basic principles of head and neck tumors
and reconstruction. Bill has published 186 peerreviewed articles, 65 chapters, and 3 texts, including the classics The Paraganglionic Chemoreceptor
System: Physiology, Pathology, and Clinical Medicine (Zak, Lawson) and Surgery of the Paranasal
Sinuses (Blitzer, Lawson, Friedman). I am honored
to have Bill as my guest of honor, recognizing all the
tireless efforts he made in teaching me, and as my
friend for over 35 years.
19
PRESENTATION OF CHEVALIER JACKSON AWARD TO
CLARENCE T. SASAKI, MD
ANDREW BLITZER, MD, DDS
Clarence Sasaki was born in Hawaii and attended
college in California. He migrated east and received
his MD from Yale University. He did general surgery
at Dartmouth Medical Center, served in the military
in Vietnam, and then returned to Yale for otolaryngology residency training. He has remained a loyal
faculty member and major pillar of the institution.
He is recognized worldwide for his contribution to
the basic and clinical science of laryngology. He has
given numerous named lectureships; has received
numerous prizes for his contributions, including the
Keese Prize from Yale, the Fowler Award from the
Triologic Society, the Casselberry Award from the
American Laryngological Association, the BroylesMaloney Award from the ABEA, the Chevalier
Jackson Lecturer for the ABEA, and the Guest of
Honor and Presidential Citation from the American
Laryngological Association. He is a member of 11
editorial boards and is a research advisor or investigator to over 30 scientific projects. He is a member
of over 20 professional organizations and has held
various offices of 7 of them, including President of
the ABEA and the Dysphagia Research Society. His
scholarly contributions are acknowledged worldwide and include 214 peer-reviewed articles, 68
chapters, and 7 textbooks. He is the quintessential
scholar, scientist, and teacher, and we are fortunate
to have him as part of the ABEA. I am honored and
privileged to call him my friend.
20
GUESTS OF HONOR 1951-2010
1951
1959
1961
1962
1963
1964
1965
1966
1967
1968
1969
1970
1971
1972
1973
1974
1975
1976
1977
1978
1979
1980
1981
1982
1983
1984
1985
FERNAND EEMAN, MD
LOUIS H. CLERF, MD
W. LIKELY SIMPSON, MD
EDWIN N. BROYLES, MD
SAM E. ROBERTS, MD
LYMAN RICHARDS, MD
VERLING K. HART, MD
JULIUS W. MCCALL, MD
FRANCIS W. DAVISON, MD
DEAN M. LIERLE, MD
LEROY A. SCHALL, MD
HERMAN J. MOERSCH, MD
LOUIS H. CLERF, MD
JOSEPH P. ATKINS, MD
RICARDO TAPIA ACUNA, MD
PAUL H. HOLINGER, MD
ARTHUR M. OLSEN, MD
FRANCIS E. LEJEUNE, SR, MD
ALDEN H. MILLER, MD
CHARLES M. NORRIS, MD
CHARLES F. FERGUSON, MD
EMILY LOIS VAN LOON, MD
DONALD F. PROCTOR, MD
FRANK D. LATHROP, MD
JOHN E. BORDLEY, MD
GABRIEL F. TUCKER, JR, MD
STANTON A. FRIEDBERG, MD
1986
1987
1988
1989
1990
1991
1992
1993
1994
1995
1996
1997
1998
1999
2000
2001
2002
2003
2004
2005
2006
2007
2008
F. JOHNSON PUTNEY, MD
HOWARD A. ANDERSEN, MD
JOHN P. FRAZER, MD
PAUL H. WARD, MD
D. THANE R. CODY, MD
M. STUART STRONG, MD
BRUCE BENJAMIN, FRACS
DAVID R. SANDERSON, MD
MICHAEL E. JOHNS, MD
JOHN A. KIRCHNER, MD
ROBERT W. CANTRELL, MD
EIJI YANAGISAWA, MD
LAUREN D. HOLINGER, MD
WILLIAM R. HUDSON, MD
ROBERT H. OSSOFF, DMD, MD
TREVOR J. MCGILL, MD
FLAVIO APRIGLIANO, MD
STANLEY M. SHAPSHAY, MD
MINORU HIRANO, MD, PHD
R. ROX ANDERSON, MD
HUGH F. BILLER, MD
FRANK E. LUCENTE, MD
MARVIN P. FRIED, MD
MARSHALL STROME, MD
2009 JAMES PEPA
2010 WILLIAM LAWSON, MD
RECIPIENTS OF CHEVALIER JACKSON AWARD
(Established 1958)
1959
1960
1961
1962
1963
1964
1965
1966
1967
1968
1969
1970
1971
1972
1973
1974
1975
1976
1977
1978
1979
1980
1981
1982
1983
1984
LOUIS H. CLERF, MD
(No award)
HERMAN J. MOERSCH, MD
PAUL H. HOLINGER, MD
EDWIN N. BROYLES, MD
LEROY A. SCHALL, MD
HERBERT W. SCHMIDT, MD
PAUL G. BUNKER, MD
JOEL J. PRESSMAN, MD
VERLING K. HART, MD
JOSEPH P. ATKINS, MD
ANDERSON C. HILDING, MD
ROBERT M. LUKENS, MD
CHARLES M. NORRIS, MD
ARTHUR M. OLSEN, MD
CHARLES F. FERGUSON, MD
SHIGETO IKEDA, MD
BLAIR FEARON, MD
FRANCIS W. DAVISON, MD
SEYMOUR R. COHEN, MD
M. STUART STRONG, MD
DEGRAAF WOODMAN, MD
ALBERT H. ANDREWS, JR, MD
GABRIEL F. TUCKER, JR, MD
HOWARD A. ANDERSEN, MD
PAUL H. WARD, MD
1985
1986
1987
1988
1989
1990
1991
1992
1993
1994
1995
1996
1997
1998
1999
2000
2001
2002
2003
2004
2005
2006
2007
2008
2009
2010
21
BRUCE BENJAMIN, FRACS
LORING W. PRATT, MD
ROBERT S. FONTANA, MD
CHARLES W. CUMMINGS, MD
BERNARD R. MARSH, MD
DAVID R. SANDERSON, MD
WILLIAM W. MONTGOMERY, MD
JOHN A. TUCKER, MD
GERALD B. HEALY, MD
VINCENT J. HYAMS, MD
LAUREN D. HOLINGER, MD
STANLEY M. SHAPSHAY, MD
ROBERT H. OSSOFF, DMD, MD
JOHN M. FREDRICKSON, MD
HASKINS KASHIMA, MD
EIJI YANAGISAWA, MD
WILLIAM W. MONTGOMERY, MD
JACK L. GLUCKMAN, MD
ELLEN M. FRIEDMAN, MD
ROBIN T. COTTON, MD
CHARLES W. VAUGHAN, MD
ANDREW BLITZER, MD
GAYLE E. WOODSON, MD
ROBERT J. TOOHILL, MD
PETER KOLTAI, MD
CLARENCE T. SASAKI, MD
INTRODUCTION OF CHEVALIER JACKSON LECTURER
MARSHALL STROME, MD
ANDREW BLITZER, MD, DDS
Marshall Strome grew up in Michigan, went to
school at the University of Michigan, and completed his otolaryngology residency at the University of
Michigan. He served briefly on the faculty of the
University of Connecticut, and then joined the faculty of the Beth Israel Hospital, Brigham and Women’s Hospital, and Harvard Medical School, where
he served for over 20 years as Chief of Otolaryngology. In 1993 he was recruited to be the Professor
and Chairman of the Otolaryngology Department
of the Cleveland Clinic, where he served for 11
years. He is now a major part of the New York Head
and Neck Institute and is a member of the faculty
at the New York Center for Voice and Swallowing
Disorders, teaching our fellows endoscopic surgery
for cancer of the larynx and pharynx. Marshall has
received countless awards and visiting professorships from around the world for his work, including
the Medal of the City of Paris; Medallion of Honor
from the American Academy of Facial Plastic and
Reconstructive Surgery; the Sword of Saudi Arabia;
and the Millenium Ogura Lecturer. He has been an
officer for many of our societies, including President of the Society of Urologic Oncology and the
American Laryngological Association. He serves on
over a dozen editorial boards. He has published over
170 peer-reviewed papers, 50 chapters, and 4 books.
His research and teaching contributions continue
to stimulate so many in our field. His novel contri-
butions include the first and only human laryngeal
transplantation, the first robotic laser surgery for a
laryngeal malignancy, and new endoscopic laryngeal preservation operations for malignancy. Most of
all, Marshall has been a ski partner and dear friend
for better than 25 years.
22
CHEVALIER JACKSON LECTURES 1964-2010
1964 DONALD F. N. HARRISON, MD. Esophageal Replacement in Postcricoid Cervical Esophageal Carcinoma.
1965 ERIC CARLENS, MD. Mediastinoscopy.
1966 JOHN L. POOL, MD (Moderator). Carcinoma of the Bronchus: A Panel Discussion.
Presentations: ERNEST L. WYNDER, MD. The Epidemiology of Cancer of the Bronchus — Facts and Suppositions.
PAUL H. HOLINGER, MD. Postsurgical Endoscopic Management of Tracheal and Bronchial Resection and
Anastomosis.
LESLIE BERNSTEIN, MD. Two Thousand Bronchoscopies in Search of Cancer.
JOHN L. POOL, MD. Is Surgery Worthwhile in Carcinoma of the Bronchus?
ROBERT H. SAGERMAN, MD. New Directions in the Radiotherapy of Lung Cancer.
1967 EELCO HUIZINGA, MD. The Tussive Squeeze and Bechic Blast of Chevalier Jackson.
1968 PAUL H. HOLINGER, MD. Management of Esophageal Lesions Caused by Chemical Burns.
1969 PLINIO DE MATTOS BARRETTO, MD. Endoscopic Surgery of the Pharyngo-esophageal Segment.
1970 JAMES R. JUDE, MD. Otolaryngological Aspects of Cardiac Arrest.
1971 JO ONO, MD. Endoscopic Microsurgery of the Larynx.
1972 G. GORDON MCHARDY, MD. Reflux Esophagitis.
1973 HERMES C. GRILLO, MD. Obstructive Lesions of the Trachea and Their Management.
1974 JOHN R. GUTELIUS, MD. Effect of Government Payment of Medical Fees on Medical Practice and Education in
Canada.
1975 DONALD O. CASTELL, MD (Captain, MC USN). Physiology and Pathophysiology of the Gastroesophageal
Sphincter.
1976 G. PAUL MOORE, PHD. Observations on Laryngeal Disease, Laryngeal Behavior and Voice.
1977 MARY ELLEN AVERY, MD. In Quest of the Prevention of Hyaline Membrane Disease.
1978 GEORGE BERCI, MD. Analysis of New Optical Systems in Bronchoesophagology.
1979 GABRIEL F. TUCKER, JR, MD. Pediatric Laryngobronchoesophagology.
1980 FLAVIO APRIGLIANO, MD. Esophageal Stenosis in Children.
1981 PETER STRADLING, MD. A Photographer’s View of the Tracheobronchial Tree.
1982 ARTHUR M. OLSEN, MD. Esophagology: An Update.
1983 BRUCE BENJAMIN, FRACS. Congenital Laryngeal Webs.
1984 RONAN O’RAHILLY, MD. Respiratory and Alimentary Relations in Staged Human Embryos. New Embryological
Data and Congenital Anomalies.
1985 JOHN A. TUCKER, MD (Moderator). Esophageal Reflux in Children: A Panel Discussion. Participants: Samuel R.
Schuster, MD; Sandra Sue Kramer, MD; Marvin Ament, MD.
1986 WILLIAM G. ANLYAN, MD. Critical Issues in Medicine Today.
1987 GUI-YI TU, MD. Stomach Pull-up and Colon Transfer in the Management of Cancer of the Hypopharynx and
Cervical Esophagus.
1988 LUCIUS D. HILL III, MD. What Would Chevalier Jackson Think?
1989 BERNARD R. MARSH, MD. Teaching Bronchoesophagology in Otolaryngology–Head and Neck Surgery Residencies in the United States.
1990 DAVID R. SANDERSON, MD. Health-Care Economics in the 1990s.
1991 MICHAEL E. JOHNS, MD. If I Had to Do It Again Would I Want It to Be the Same as Before?
1992 WHITNEY A. ADDINGTON, MD. Needed: The Reorganization of Health Care, Not More Money.
1993 HENRY J. HEIMLICH, MD, SCD. The Heimlich Maneuver in Infants and Children: The Best Treatment for Saving
Drowning and Choking Victims.
1994 JOHN A. KIRCHNER, MD. The Glottic Gateway.
1995 MINORU HIRANO, MD. Phonosurgery: Past, Present, and Future.
1996 HAROLD C. PILLSBURY III, MD. A Strategy for Academic Specialists in a Managed Care World.
1997 GERALD B. HEALY, MD. The Legacy of Jackson: Has It Been Lost?
1998 ROBIN T. COTTON, MD. The Story of Laryngotracheal Reconstruction in Children.
1999 JAMES A. KOUFMAN, MD. Laryngopharyngeal Reflux: A New Paradigm of Airway Disease.
2000 STANLEY M. SHAPSHAY, MD. Twenty-Five Years’ Experience With Endoscopic Laser Treatment for Vocal Cord
Cancer: What Is the Standard of Care in Y2K?
2001 PAUL A. LEVINE, MD. Accurate Endoscopic and Radiologic Pretreatment Evaluation: The Jackson Legacy and Its
Impact on the Evolution of Therapy for Sinonasal Malignancies.
2002 STEVEN D. GRAY, MD. Tissue Engineering the Laryngeal Components: Pushing the Edges of Biotechnology.
2003 WOLFGANG STEINER, MD. Transoral Surgical Management of Hypopharyngeal Carcinoma.
2004 JONATHAN E. AVIV, MD. Transnasal Esophagoscopy: From New Frontier to the Future of Esophagology.
2005 JOHN L. WARD, PHD. Cancer of the Vocal Folds: A Patient’s Perspective.
2006 STEVEN M. ZEITELS, MD. Concepts and Culture of Innovation.
2007 PEAK WOO, MD. Office-Based Interventional Laryngology: Our Legacy and Our Future.
2008 REZA SHAKER, MD. Reciprocal Physiology and Pathophysiology of the Upper Gut and Airway.
2009 CLARENCE T. SASAKI, MD. The Vagus Nerve: Dispelling the Animal Spirits.
2010 MARSHALL STROME, MD. Laryngeal Transplantation: The End of the Beginning.
23
PRESIDENTIAL CITATIONS HONORING
ANTHONY F. JAHN, MD, MITCHELL F. BRIN, MD,
AND ROBERT M. BLITZER, DDS
ANDREW BLITZER, MD, DDS
ANTHONY F. JAHN (L) & ANDREW BLITZER (R)
Born in Budapest and schooled in Toronto, Anthony F. Jahn is the product of a University of Toronto residency. He comes from professional music
family and is himself a concert pianist. I met Tony
in 1979, when he joined our faculty as an otologist
at Columbia. In a short time, he also worked with
Jim Gould and Eugene Grabscheid in treating professional voice. He joined the staff of doctors at the
Metropolitan Opera Company, and recently became
the medical director of the Metropolitan Opera Company, consultant to the Juilliard School of Music,
consultant laryngologist for the Royal Shakespeare
Company, and Adjunct Professor of Voice Pedagogy
at the Westminster Choir College in Princeton, New
Jersey. Tony has served as Professor and Chairman
of Otolaryngology at the University of Medicine
and Dentistry of New Jersey, and now serves as Professor of Clinical Otolaryngology of the Columbia
University College of Physicians and Surgeons. He
teaches the care of professional voice to our fellows
at the New York Center for Voice and Swallowing
Disorders.
He has given lectures and taught courses worldwide for greater than 30 years on otology and voice.
He is a trained and certified acupuncturist and a student of Eastern medicine. He has for many years
participated in mercy medical missions to southeast
Asia. He has been a guest examiner of the American Board of Otolaryngology, serves as a reviewer
or editorial board member for a dozen journals, and
has written over 90 peer-reviewed articles, chapters,
or books, including the classic Care of the Professional Voice. Most importantly, Tony has been a best
friend for over 30 years.
24
Presidential Citations
25
ANDREW BLITZER (L) & MITCHELL F. BRIN (R)
Mitchell F. Brin grew up in New Jersey, and went
to college at the University of Pennsylvania. He returned to New York and was a student at Columbia College of Physicians and Surgeons. I first met
Mitchell as a third-year student and tried to convince
him to go into otolaryngology. He chose neurology
because he didn’t like having to get to the operating room so early in the morning, only to find out
years later that he was seeing patients at 7 AM. He
did his internal medicine at Mt Sinai, neurology at
Columbia, and his movement disorders neurology
fellowship at Columbia. He then became part of the
Stan Fahn Movement Disorders Center at Columbia,
where I began working with him on head and neck
dystonias and botulinum toxin therapy. Mitchell and
I worked together closely in treating a variety of
neurologic conditions affecting the larynx, including spasmodic dysphonia, Parkinson’s disease, and
tremor. Mitchell worked at Columbia until 1994,
when he moved to the Mt Sinai Medical Center in
New York, where he is Bachman-Strauss Professor
of Movement Disorders Neurology. He currently is
also Vice-President for Development and Therapeu-
tic Head, Botox Neurology Worldwide for Allergan
Pharmaceutics. Among Mitchell’s many talents are
his keen research analytical abilities. He has served
as a scientific advisor to the Parkinson Study Group,
American Association of Neurology Therapeutics
subcommittee, National Spasmodic Dysphonia Association, founder of We Move, founder of the Soviet-American Neurogenetics Group, review board
member of US Pharmacopeia, Associate Member
of the American Academy of Otolaryngology–Head
and Neck Surgery, and member of the Neurolaryngology Study Group, among many others. He has
taught worldwide on evaluation and treatment of
movement disorders. He serves as a reviewer for 15
journals, as well as an ad hoc reviewer for the National Institutes of Health. He has written over 100
peer-reviewed papers, 60 chapters, 116 abstracts or
reviews, and 50 collaborative papers from the Parkinson Study Group, and is a coeditor for our text
Neurologic Disorders of the Larynx, now in its second edition. Most of all, he has been a dear friend
for over 30 years.
26
Presidential Citations
ROBERT M. BLITZER (L) & ANDREW BLITZER (R)
Robert M. Blitzer, whom I have known longer
than all of the other award winners, was born in New
York and schooled in New York. He received his
DDS degree from Temple University, where he became board-certified in prosthodontics and was part
of the dental faculty until 1995. During his tenure
at Temple, he became an expert in the science and
evolution of dental materials. His research led him
to become a consultant and then in 1995 director
of Clinical Research for Dentsply in Delaware. He
continued the clinical practice of dentistry, as well
as teaching. He is an Associate Professor of Dentistry at the University of Maryland as has served
as a consultant to the Wilkes-Barre VA and the VA
Medical and Regional Office Center in Wilmington, Delaware. He has expanded the evaluation and
treatment of dysphagia (particularly management of
oral phase deficits with prosthetic dental devices and
swallowing retraining). He has lectured worldwide,
and his science continues to be used in the innovation of new materials. He is the recipient of honors
and invited lectureships worldwide. He has numerous publications and has received many honors as
an outstanding teacher. Most of all, he is my baby
brother whom I love.
PRESIDENT-ELECT ADDRESS
MICHAEL A. ROTHSCHILD, MD
Standing on the shoulders of giants makes any
job easier, and this one is no exception. In my first
official communication, I have to acknowledge my
predecessors in this role — from Dr Jackson to Dr
Blitzer — who have done so much to make the
ABEA into the renowned and scholarly association
that it is today.
Over the next 12 months, we will be implementing several initiatives launched during the last administration. We will be accepting applications for
the new resident member class (with a representative
at the board meeting). Also, we will be developing
the Past Presidents council that will spearhead the
development effort begun by Dr Sasaki and serve
as a source of advice and perspective in the years
to come. We are in the process of bringing a number of current members into leadership roles on the
council, and we continue our ongoing membership
drive, bringing new colleagues in the field of bronchoesophagology into our ranks.
I am looking forward to a productive year, culminating in our annual spring meeting. I hope to see
you all in Chicago in 2011, where I am anticipating yet another terrific forum for the presentation of
new research, and the renewal of old friendships.
27
ABEA Panel on Laryngopharyngeal Reflux
Moderator: Peter Belafsky, MD, PhD
Panelists: Jamie Koufman, MD; Tanya Meyer, MD;
Nikki Johnston, PhD; Michael Pitman, MD
Case Presentation. Thirty-eight-year-old man
with chief complaint of globus failing treatment
with twice-daily proton pump inhibitors. Also with
symptoms of mild dysphagia and excessive throatclearing (Reflux Symptom Index [RSI] = 18, Eating
Assessment Tool-10 [EAT-10] = 9).
Panel Recommended Subsequent Evaluation and
Treatment. Balloon dilation of the upper esophageal
sphincter to address the cricopharyngeal bar as a
treatment of his globus and a dilation of his Schatzki
ring as a treatment of his dysphagia. The question
was raised about whether the Schatzki ring could be
protective against reflux. The consensus from the
panel was that most gastroesophageal reflux is from
transient lower esophageal sphincter relaxations and
that dilating the ring would not make the gastroesophageal reflux worse.
Pharmaceutical History. Hypertension on Prinizide.
Social History. Attorney and professional voice
user. No tobacco or illicit drugs. Social alcohol use.
Panel Recommended Evaluation. Comprehensive
head and neck examination with flexible video laryngoscopy.
Results of Dilation to 60F of Upper Esophageal
Sphincter and Schatzki Ring. Improved dysphagia
(EAT-10 = 4). No improvement in globus or throatclearing.
Results of Initial Evaluation. Normal head and
neck examination with evidence of chronic laryngitis on laryngoscopy with vocal fold edema and an
early granuloma formation (Reflux Finding Score
[RFS] = 9).
Panel Recommended Subsequent Evaluation and
Treatment. Twenty-four hour dual-probe ambulatory pH and impedance testing without proton pump
inhibitors.
Panel Recommended Subsequent Evaluation.
Unsedated transnasal esophagoscopy.
Results of pH and Impedance Testing. The patient’s symptoms of globus and throat-clearing got
worse after abruptly stopping the twice-daily proton
pump inhibitors. The reflux testing revealed that the
pH was less than 4 for 3.6% of the time. There were
3 episodes of a pH less than 4 in the hypopharynx, 5
episodes of a pH between 4 and 5 in the hypopharynx, and 14 weakly-acid reflux episodes. Negative
symptom association with throat-clearing.
Results of Transnasal Esophagoscopy. Mildly
constricting Schatzki B ring. No evidence of peptic
esophagitis or Barrett metaplasia. It was felt by the
panel that the Schatzki ring was potentially the cause
of the patient’s dysphagia but unlikely the cause of
his globus.
Panel Recommended Subsequent Evaluation and
Treatment. The patient should be offered simple reassurance vs. a further work-up with fluoroscopy
and manometry. The patient remained significantly
bothered by the globus and desired a more-extensive
work-up. A fluoroscopic swallow study and highresolution pharyngeal and esophageal manometry
was performed.
Panel Recommended Subsequent Evaluation and
Treatment. Because of the abnormal pH testing, the
patient was placed back on proton pump inhibitors,
and a strict low-acid diet, liquid alginate 30 mL after meals and before bedtime, and baclofen 20 mg
3 times per day were recommended. Because of the
relationship between ACE inhibitors and cough and
the potential relationship between ACE inhibitors
and throat-clearing, the patient was referred back
to his primary care physician to discuss replacing
the ACE inhibitors with another anti-hypertensive
medication.
Results of Fluoroscopy. Non-obstructing cricopharyngeal bar, 2-cm hiatal hernia, and a mildly
constricting Schatzki B ring.
Results of Manometry. Normal pharyngeal peak
pressure with slightly elevated upper esophageal
sphincter residual pressure. Normal esophageal
body motility.
Results of Aforementioned Treatment. Globus
and throat-clearing significantly improved. RSI = 7
(within normal limits).
28
SPECIAL PRESIDENTIAL LECTURE
Airway Clinic: Bronchoesophagology in the Twenty-First Century
Marshall E. Smith, MD; Mark Elstad, MD
By nature of the anatomical overlap of the upper aerodigestive tract, diseases of these structures affect a variety of medical
specialties. Team care for clinical conditions that cross specialty boundaries has become the standard of care for problems
such as clefts and craniofacial disorders in pediatrics and head and neck cancer in adults. In diagnosis and treatment of
diseases of the esophagus and tracheobronchial tree, the specialties of pulmonary medicine and gastroenterology have also
made significant advances in diagnosis and treatment in the past 30 years. This report describes our 12-year experience
with a multidisciplinary clinic for team care management of diseases of airway obstruction in adults. An update from the
emerging field of interventional pulmonology is highlighted.
We thank President Blitzer, Guests of Honor, and
members of the ABEA for the opportunity to present
our perspective on a multidisciplinary approach to
bronchoesophagology. Our theme is that collaborative care wins — for patients and the caregivers. This
type of model for clinical care is not totally novel.
Many members of the ABEA are pediatric otolaryngologists. They know that in children’s hospitals,
care teams have evolved for decades in treatment
of conditions such as clefts and craniofacial disorders and spina bifida. The internet was surveyed to
find pediatric hospitals that have a team for care of
aerodigestive tract disorders. There are many programs now in place for managing airway, voice, and
swallowing disorders in children. The teams vary
in composition. They include otolaryngology, pulmonary medicine, gastroenterology, sleep medicine,
pediatric surgery, speech-language pathology, respiratory therapy, and nursing. Some have scheduled
care conferences, and some have immediate care
coordination during the clinic visit. A key feature
of collaborative care is that the patient sees more
than one specialist during a clinic visit, and the specialists discuss and coordinate each patient’s care.
In head and neck cancer care, team management
has also become standard through collaboration between specialists in head and neck surgery, radiation
oncology, medical oncology, reconstructive surgery,
dentistry, prosthodontics, prosthetics, pathology, radiology, speech-language pathology, and nursing.
strengths and resources in their particular institution
and situation and build them up. At the University
of Utah, the airway clinic’s origins can be traced to
the early 1990s, when Dr Steve Gray, a prominent
member of the ABEA, was working on a particularly difficult case of laryngotracheal papillomatosis.
He asked Dr Elstad to help him manage the disease
in the lower airway. Dr Elstad was doing rigid bronchoscopy as a self-taught interventional pulmonologist, treating mainly endobronchial tumors as he
learned from a text by Dr Chevalier Jackson1 and
observation of Dr Jim Harrell. Drs Gray and Elstad
also collaborated in caring for some patients with
airway stenosis with endotracheal stents.2 When Dr
Smith came to the University of Utah in 1997, the
airway patients came to the voice disorders clinic.
This was inefficient for the speech pathologist, and a
pulmonologist was not available for the complicated
cases. Drs Smith and Elstad decided that it would
be far better to have patients who required more
than one specialist to be scheduled at the same time.
They had enough patients that they began to run a
separate clinic. In 1998, they opened the University
of Utah Airway Disorders Clinic and began seeing
patients together routinely. For 3 years, a gastroenterology fellow worked there, too, making for a full
aerodigestive team. The clinic is currently working
to recruit another interested gastroenterologist. At
the Airway Clinic, there are 2 half-days each month
when patients are jointly seen in dual, 30-minute
appointments with both the laryngologist and the
pulmonologist. Patients are seen in an outpatient
Each bronchoesophagologist needs to assess the
From the Division of Otolaryngology–Head and Neck Surgery (Smith) and the Division of Critical Care, Occupational, and Pulmonary
Medicine, Department of Internal Medicine (Elstad), University of Utah School of Medicine, Salt Lake City, Utah.
Correspondence: Marshall E. Smith, MD, Division of Otolaryngology–Head and Neck Surgery, University of Utah School of Medicine,
3C-120 SOM, 50 N Medical Dr, Salt Lake City, UT 84132.
29
30
Special Presidential Lecture
otolaryngology clinic in rooms that are equipped
for endoscopy. They are examined, the endoscopic
examinations are recorded, and then the cases are
staffed together by both doctors.
The most common disorders seen are idiopathic
subglottic stenosis and postintubation stenosis. The
clinic sees a few adults with problems from congenital diseases. Other conditions include autoimmune
diseases affecting the airway, tracheobronchomalacia, laryngeal paralysis, vocal fold paralysis and fixation, and occasionally, anastomotic stenosis from
lung transplantation.
For evaluation, the clinic relies heavily on the laryngotracheobronchoscopy procedure.3 This is the
diagnostic procedure that makes the Airway Clinic
work. It is done awake and unsedated and accomplishes the assessment in almost all patients. No
separate sedated bronchoscopy is needed. It helps
considerably with presurgical planning. Training and
expertise in this procedure is a crucial component of
the success of the Airway Clinic. This procedure can
be learned through courses at the AAO-HNS annual
meeting, which are generally taught by members of
the ABEA. Both otolaryngologists and pulmonologists should develop skill with this procedure. Ideally, the examinations should be done jointly, so
technical facility can be compared and constructively critiqued. About 60% of the bronchoscopy procedures performed at the clinic are for evaluation
and diagnosis, and about 40% are for treatment. The
speed of the procedure is a big advantage. Outpatient flexible bronchoscopy is performed in minutes
instead of the typical 2 hours when done under sedation in the conventional pulmonologist endoscopy
clinic. Thus, it is a very efficient method for evaluating the airways.
The activities of the Airway Clinic also include
surgical treatment and evaluation of outcomes. Examples of studies at our center include the effect of
mitomycin C treatment of laryngotracheal stenosis,
and the vocal consequences of cricotracheal sleeve
resection.4,5 In 2009, the operative logs of the clinic included 14 open reconstructive procedures, 75
endoscopic procedures, 10 stent placements, 9 intrabronchial valve replacements, and 20 diagnostic
bronchoscopies with sedation. One of the newer
bronchoscopic procedures being used at the clinic
is endobronchial ultrasound for diagnosis of bronchogenic carcinoma. This procedure has essentially
eliminated the need for mediastinoscopy in evaluating the disease. Intrabronchial valves are a new device for treating emphysema patients. Lung volume
reduction surgery has resulted in improvement in
both patient quality of life and measurable gains in
pulmonary function. However, it is an expensive and
often morbid procedure. A trial is now under way to
assess intrabronchial valves that are placed through
a bronchoscope into the segmental bronchial airways. They self-expand and contract and redirect air
to healthier tissue. This is a less-invasive alternative
that may achieve similar results. Our clinic is participating in the device trial and looking at patient
satisfaction and survival rates.
In summary, the combined resources and expertise of laryngology and pulmonology in evaluation
and management of airway lesions yield many positive benefits for patients and clinicians. Patients frequently come to the clinic complaining of shortness
of breath, and it’s not always due to an airway lesion, even though a known airway lesion may exist.
Other causes of dyspnea, related to different organ
systems, are important to recognize, and the benefit
of having a physician with an internal medicine perspective to help assess and manage them is invaluable. Collaboration with doctors in other specialties
is as simple as going ahead and doing it. Those interested in developing a clinical practice model like this
can find appropriate colleagues to develop expertise
with and simply start seeing patients together.
REFERENCES
1. Jackson C, Jackson CL. Bronchoesophagology. Philadelphia, Pa: WB Saunders, 1950.
2. Kurrus JA, Gray SD, Elstad MR. Use of silicone stents in
the management of subglottic stenosis. Laryngoscope 1997;107:
1553-8.
3. Hogikyan ND. Transnasal endoscopic examination of the
subglottis and trachea using topical anesthesia in the otolaryn-
gology clinic. Laryngoscope 1999;109:1170-3.
4. Smith ME, Elstad M. Mitomycin C and the endoscopic
treatment of laryngotracheal stenosis: are two applications better than one? Laryngoscope 2009;119:272-83.
5. Smith ME, Roy N, Stoddard K, Barton M. How does cricotracheal resection affect the female voice? Ann Otol Rhinol
Laryngol 2008;117:85-9.
Arytenoid Abduction: Indications and Limitations
Gayle Woodson, MD
Objectives: I report further experience with arytenoid abduction (AAb), a procedure that enlarges the glottis by external
rotation of the arytenoid cartilage and thus moves the vocal process laterally and rostrally, but does not preclude adduction
for phonation. Therefore, AAb has the potential to preserve voice in patients with bilateral abductor laryngeal paralysis.
Methods: I performed a retrospective review of AAb in 11 patients with bilateral laryngeal paralysis and 3 patients with
other neurologic causes of glottal airway compromise, ie, adductor breathing dystonia, frequent laryngospasm, and progressive laryngeal breathing dysfunction.
Results: Seven of the 11 patients with bilateral paralysis had dramatic airway improvement. One patient required a tracheotomy after AAb, and 3 patients with an existing tracheotomy could not be decannulated. Arytenoid abduction relieved airway obstruction in the patient with recurrent laryngospasm and in the child with progressive laryngeal breathing
dysfunction, but the patient with adductor breathing dystonia has persistent stridor. The factors associated with a poor airway outcome included prolonged tracheotomy, electromyographic evidence of inspiratory activity of adductor muscles,
chronic obstructive pulmonary disease, sleep apnea, and prior cordotomy or arytenoidectomy.
Conclusions: Arytenoid abduction is most effective in patients with bilateral laryngeal paralysis of less than 1 year’s duration who do not have unfavorable laryngeal adductor activity.
Key Words: arytenoid cartilage, bilateral laryngeal paralysis, surgery.
are temporary, and the injection must be repeated
every few months. All of these approaches not only
enlarge the airway, but also increase the glottal gap
during phonation. As a result, the voice is weaker
and breathier, and speech requires more effort. Every incremental gain in breathing is accompanied by
further loss of voice. Sometimes, patients have significant aspiration and recurrent pneumonia because
the glottis does not close during the swallow.9,10 The
optimal result expected from these procedures is a
compromise, to improve breathing without causing
aspiration during swallowing or an unacceptable deterioration of voice.11,12
A better functional result could be achieved if dynamic function could be restored, with opening for
breathing and closing for phonation. Through the
years, much effort has focused on surgical reinnervation to restore movement to the paralyzed larynx.
However, to date, only limited success has been reported.13-16
The arytenoid abduction (AAb) procedure is intended to enlarge the airway in patients with bilateral laryngeal paralysis while preserving residual
or recovered motion in adductor muscles. Clinical
experience and animal experiments indicate that af-
INTRODUCTION
Bilateral vocal fold immobility is one of the most
challenging clinical problems encountered in the
practice of laryngology. The primary symptom is airway obstruction, as the vocal folds do not open during inspiration. Patients also have various degrees
of vocal impairment. Tracheotomy provides an adequate airway, and does not impair glottal function in
phonation. However, a tracheotomy is a significant
functional and cosmetic handicap. The first surgical
procedures used to relieve airway obstruction in patients with bilateral laryngeal paralysis were external approaches to cordectomy, arytenoidectomy, and
arytenopexy.1-3 Endoscopic approaches were also
described,4 and with the advent of laser surgery, endoscopic arytenoidectomy, cordectomy, and cordotomy became technically easier.5,6 A vocal fold lateralization suture is particularly useful in patients with
acute laryngeal paralysis, in whom there is a possibility that motion could be recovered.7 However, all
of these procedures improve breathing by statically
enlarging the glottal airway. Botulinum toxin injection into adductor muscles is another option that has
been reported to improve the airway in these patients, with variable results.8 The effects of the toxin
From the Division of Otolaryngology, Southern Illinois University School of Medicine, Springfield, Illinois.
Presented at the meeting of the American Broncho-Esophagological Association, Las Vegas, Nevada, April 28-29, 2010.
Correspondence: Gayle Woodson, MD, Division of Otolaryngology, Southern Illinois University School of Medicine, Springfield, IL
62794-9662.
31
32
Woodson, Arytenoid Abduction Revisited
A
B
Fig 1. (Patient 11) Direct laryngoscopic view of larynx of patient 11 demonstrates passive mobility of arytenoid
cartilages. A) Vocal folds at rest. B) Vocal folds displaced by tip of laryngoscope.
ter recurrent laryngeal nerve injury, the vocal fold
gradually migrates to a midline position.17-19 This
change in position has been attributed to preferential
reinnervation of adductor muscles.19,20 With little or
no reinnervation of the abductor muscles, the vocal
folds remain in the adducted position. The AAb procedure was designed on the basis of 3-dimensional
motion analysis research in cadaver larynges that
indicated that the posterior cricoarytenoid (PCA)
muscle, the only abductor muscle of the larynx,
and the adductor muscles rotate the arytenoid cartilage around different axes.21 Furthermore, when
an arytenoid adduction suture is in place, an AAb
suture does not move the vocal process away from
the midline.22 Endoscopic observations in patients
undergoing arytenoid adduction surgery have confirmed that the vocal process can be medialized by
an arytenoid adduction suture, even during traction
on a second suture that mimics the action of the PCA
muscle.22
On the basis of these observations, the AAb procedure was designed. It uses a single suture to pull
the muscular process of the arytenoid cartilage inferiorly. This externally rotates the arytenoid cartilage
to move the vocal process laterally and superiorly,
to enlarge the glottis.23 A distinction must be made
between bilateral vocal fold immobility due to mechanical fixation and paralysis due to neural impairment of laryngeal muscles. In patients with paralysis, the vocal folds can be passively displaced laterally (Fig 1); however, when there is fixation of the
vocal folds or the cricoarytenoid joint by scar tissue,
AAb would not be effective.
Arytenoid abduction was previously reported as
a promising surgical procedure that reduced airway
obstruction without sacrificing vocal function in 6
patients with bilateral laryngeal paralysis.23 Further
experience with this treatment for a relatively uncommon condition is presented to clarify the indications for the procedure and to review unsuccessful
cases, so that factors that may have contributed to
unsatisfactory results may be identified.
METHODS
The records of the Southern Illinois University
Voice Center from June 2005 through December
2009 were reviewed to identify patients in whom
AAb had been performed. Each patient had presented with airway obstruction and bilateral vocal
fold immobility and had a diagnosis made by videoendoscopy in the clinic. Direct laryngoscopy was
performed under general anesthesia to assess the
passive mobility of the arytenoid cartilages. Lateral pressure was applied on the membranous vocal
fold to displace it while the arytenoid cartilage was
observed for motion. The vocal folds were also simultaneously displaced with the tip of a Holinger
laryngoscope (Fig 1). If the vocal folds had a good
range of motion, AAb was performed. Briefly, the
posterior larynx was exposed by the same cervical
approach used in arytenoid adduction. A suture was
placed through the muscular process, and then inferior traction was applied. The larynx was observed
endoscopically to confirm lateral displacement of
the vocal process, and then the suture was secured
to the inferior cornu of the thyroid cartilage. Patients
without a tracheotomy were extubated in the operating room after the procedure and admitted to the
hospital overnight for observation. Patients with an
existing tracheotomy were discharged on the same
Woodson, Arytenoid Abduction Revisited
33
PATIENTS WITH BILATERAL LARYNGEAL PARALYSIS WHO UNDERWENT ARYTENOID ABDUCTION
Patient
No.
Sex
Presenting
Airway
Airway
Outcome
Presenting
Voice
Voice
Outcome
Prior
Surgery
1
2
3
4
F
F
F
F
Intubation
Idiopathic
Thyroidectomy
Intubation
Stridor, dyspnea
Stridor, distress
Stridor, dyspnea
Tracheotomy
Somewhat breathy
Slightly weak
Weak, breathy
Weak, breathy
No change
Same
Slightly worse
Same
No
No
Cordectomy
Multiple
F
M
Thyroidectomy
Thyroidectomy
Stridor, dyspnea
Tracheotomy
Weak, breathy
Normal
Weaker
Same
Multiple
No
7
M
Congenital
Tracheotomy
Weak
Same
No
8
9
10
11
F
M
M
F
Thyroidectomy
Esophageal cancer
Lung cancer
Thyroidectomy
Stridor, distress
Stridor, distress
Tracheotomy
Tracheotomy
Tracheotomy
No stridor
No stridor
Better, still has
tracheotomy
Slight stridor
Better, still has
tracheotomy
Better, still has
tracheotomy
No stridor
No stridor
Decannulated
Decannulated
5
6
Normal
Weak, breathy
Weak
Weak
Normal
Same
Same
Improved
No
No
No
No
Cause
day with the tracheotomy in place, and then returned
to the clinic after a few days for a trial of decannulation.
RESULTS
BILATERAL LARYNGEAL PARALYSIS
Eleven patients with bilateral paralysis underwent
AAb. The cause of paralysis was thyroidectomy in
5 patients and idiopathic in 2 patients. Other causes
included intubation (in 2 patients), esophageal cancer, and lung cancer. Five patients presented with a
tracheotomy in place, whereas 4 had acute airway
distress and had AAb performed urgently as an alternative to tracheotomy. Two other patients had
significant restriction of activity due to stridor, and
AAb was performed electively. Three patients had
previously undergone arytenoidectomy and/or cordotomy. Patient information is detailed in the Table.
Successful Airway Outcomes. Seven patients with
bilateral paralysis had immediate and dramatic improvement in the airway, including 2 patients with a
preexisting tracheotomy who were subsequently decannulated. Figure 2 shows views of the larynx during inspiration and during phonation before and after AAb. This patient had fairly good mobility of the
left arytenoid cartilage; however, that function was
obscured by the overhanging right arytenoid cartilage. After AAb, the right arytenoid cartilage was
lifted out of the airway and the motion of the left
arytenoid cartilage was “unmasked.”
Two patients noted a slight decrease in vocal
function. One patient had previously undergone endoscopic cordotomy. That procedure had resulted in
immediate airway improvement, but over time the
patient developed recurrent airway obstruction due
to scar contracture of the glottis. Her voice was significantly breathy before AAb, and after the procedure it was noticeably more breathy. Only 1 other
patient noticed a decrease in vocal function. None
of the patients developed aspiration during swallowing.
Failures. Three of the patients with an existing
tracheotomy could not be decannulated, and patient 1, who did not originally have a tracheotomy,
had persistent stridor after AAb and eventually required tracheotomy. Laryngeal electromyography
performed in 2 of the patients with persisting airway
obstruction after AAb detected strong inspiratory activation of the thyroarytenoid (TA) muscle. In both
patients, laryngoscopy under general anesthesia had
documented mobile vocal folds, and the AAb suture
resulted in significant lateral displacement of the vocal fold under anesthesia. However, during awake
examination, both vocal folds were in the midline.
After failure of AAb, patient 1 was treated with
botulinum toxin injection into her left TA muscle.
However, her airway was not sufficiently improved
to permit decannulation. Subsequent cordotomy and
arytenoidectomy also failed to provide an accurate
airway, and tracheotomy was performed.
Patient 4, who presented with a tracheotomy, had
a complex medical history. She developed bilateral laryngeal paralysis after intubation for sepsis that
developed after a perforation during colonoscopy,
followed by pulmonary fibrosis. Tracheotomy was
performed. She was eventually decannulated, but
sought treatment for her weak voice. She underwent
left thyroplasty followed by revision thyroplasty,
and tracheotomy was performed for airway management. She presented to our clinic with the hope that
her airway could be restored. Her mean phonation
time before surgery was only 2 seconds. Larynge-
34
Woodson, Arytenoid Abduction Revisited
A
B
C
D
Fig 2. (Patient 11) Flexible laryngoscopy before and after arytenoid abduction. A) Phonation before surgery.
B) Inspiration before surgery. C) Phonation after surgery. D) Inspiration after surgery.
al stenosis was suspected because the problem had
arisen after intubation; however, at direct laryngoscopy the vocal folds were mobile to palpation, and
under anesthesia right-sided AAb significantly widened the glottis. The left thyroplasty implant was
also removed. Immediately after surgery, her airway
was markedly improved and the tracheotomy was
removed. However, 1 month later, she presented
with inspiratory stridor, and examination revealed
inspiratory adduction of the left vocal fold. Left-sided AAb was then performed, but during surgery, the
arytenoid cartilage fractured. Since surgery, she has
remained tracheotomy-dependent.
Another patient who could not be decannulated
after AAb (patient 7) was a 15-year-old boy who
had been tracheotomy-dependent nearly all his life.
After AAb, laryngoscopy revealed an improved
glottal airway, with an inspiratory glottal angle that
increased from 2° to 15°. However, he became anxious when the tracheotomy was removed or capped
and did not tolerate decannulation.
OTHER CAUSES OF GLOTTAL AIRWAY IMPAIRMENT
Arytenoid abduction was also performed in 3 patients who had glottal obstruction with a neurologic
cause other than bilateral laryngeal paralysis.
Patient 12. A 38-year-old man presented with episodic laryngospasm that had previously been managed with periodic injections of botulinum toxin.
The episodes of laryngospasm began after a tracheal
resection for acquired subglottic stenosis that had resulted from prolonged endotracheal intubation. The
botulinum toxin injections suppressed the spasms,
but resulted in a breathy voice and required frequent
repeat injection. Arytenoid abduction was offered as
an option. The procedure prevented complete obstruction during the spasm and did not impair his
voice. The patient was pleased with the ability to
maintain his airway without sacrificing his voice;
however, 1 year later he required tracheotomy because of recurrent subglottic stenosis. He currently has a T-tube in place that is plugged, so that he
breathes across the glottis. His laryngeal airway is
Woodson, Arytenoid Abduction Revisited
still adequate and is not occluded by spasm.
Patient 13. A 10-year-old boy presented to our
clinic with nocturnal airway obstruction that had
made him tracheotomy-dependent since infancy.
Evaluation at that time demonstrated inspiratory adduction of the vocal folds during sleep. He was managed with periodic botulinum toxin injection into
his TA muscles and was successfully decannulated.
Over the next 4 years, his nocturnal stridor became
less responsive to botulinum toxin and he began to
develop airway obstruction that persisted during
wakefulness. This progressed to severe stridor, despite bilateral chemodenervation of the TA muscles.
Flexible endoscopy revealed failure of inspiratory
vocal fold abduction, particularly on the left side.
Left-sided AAb was performed, and his airway was
vastly improved. Two years later, he maintains an
adequate airway without the need for botulinum toxin therapy. The cause of his progressive airway impairment is still unknown.
Patient 14. A 70-year-old man presented with inspiratory stridor. Endoscopy revealed severe edema
over the arytenoid cartilages that prolapsed into his
airway during inspiration. His vocal folds did not
abduct during quiet breathing, but opened widely
during the inspiratory phase of a cough and during
prephonatory inspiration. Electromyography demonstrated bilateral phasic inspiratory adduction of
his TA muscles, as well as episodic myotonic discharges. An electromyogram of limb muscles did
not demonstrate any evidence of systemic myopathy. He was treated with injections of as much as 30
units of botulinum toxin into the left TA muscle, and
bilateral injections of 5 units. His voice was softer
after each botulinum treatment, but his stridor persisted. He also underwent microlaryngoscopy with
excision of redundant mucosa without further improvement. After left-sided AAb, his airway and exercise tolerance were improved, but his stridor persisted. Endoscopy demonstrated inspiratory supraglottic constriction.
DISCUSSION
Bilateral laryngeal paralysis has long been managed either by tracheotomy or by surgical procedures
that statically enlarge the glottis with resection of
glottal tissue or with lateral fixation of the arytenoid
cartilage.1-7,10,11 Such glottal enlargement procedures improve the airway, but do so at the expense
of the voice, because the glottis cannot close for phonation. Additionally, there is a significant incidence
of aspiration during swallow, because laryngeal closure during swallow is impaired. The incidence of
aspiration is higher after procedures that resect the
35
body of the arytenoid cartilage.11,12 The goal of all
such procedures — arytenoidectomy, arytenopexy,
and/or cordotomy — is to gain a significant static
improvement in the airway, with an acceptable decrease in voice quality. The ideal treatment would
restore some dynamic function, so that the larynx
could open for breathing and close for speech and
swallowing. There has been much interest in reinnervation of the larynx as a means of restoring normal motion, but as yet, success is rare.12,13
Both clinical experience and animal experiments
indicate that residual adductor muscle activity is
very common in immobile vocal folds after injury
to the recurrent laryngeal nerve. This has been attributed to spontaneous regeneration of the recurrent
laryngeal nerve, with a predominance of reinnervation to adductor muscles.20 Thus, paralyzed vocal
folds tend to lie immobile near the midline, because
there is inadequate muscle force to abduct the vocal folds. This is the most likely explanation for the
classic observation that vocal folds move medially over time after recurrent laryngeal nerve injury.
Preferential reinnervation of adductor muscles and
inadequate reinnervation of abductor muscles can
account for the predominance of airway symptoms,
with fairly acceptable voice, commonly observed in
patients with bilateral paralysis. Therefore, if a procedure could provide abduction without abolishing
residual phonatory adduction, it could restore vocal
fold motion with respiration and phonation.
The AAb procedure was designed on the basis
of research in cadaver larynges that indicated that
abduction of the arytenoid cartilage does not abolish adductor motion. The cricoarytenoid joint is essentially a ball and socket, rather than a hinge joint.
Thus, motion is not restricted to simple opening and
closing. Three-dimensional motion analysis demonstrates that the force exerted by laryngeal adductor
muscles rotates the arytenoid cartilage about a nearly vertical axis. In contrast, the PCA muscle pulls
downward and medially on the muscular process,
moving the vocal process upward and outward. The
axis of rotation of the arytenoid cartilage during PCA
contraction is oblique, rather than vertical.21 Thus,
contraction of the PCA muscle does not preclude
adduction of the arytenoid cartilage by contraction
of the adductor muscles. Experiments in cadaver larynges have demonstrated that when an arytenoid
adduction suture is in place, inferior traction on the
muscular process does not move the vocal process
away from the midline.22 Endoscopic observations
in patients undergoing arytenoid adduction surgery
have confirmed that the vocal process can be medialized by an arytenoid adduction suture, even during
traction on a second suture that mimics the action of
36
Woodson, Arytenoid Abduction Revisited
the PCA muscle.
The initial experience with AAb in 6 patients resulted in excellent airway improvement with preservation of voice in 5 patients, 4 of whom had been
tracheotomy-dependent.23 In this present series,
AAb was only successful in 7 of 11 patients. Thus,
our total experience to date is that AAb significantly
improved the airway in 12 of 17 patients with bilateral laryngeal paralysis. None of the patients developed swallowing problems or aspiration.
The reported success rates for other surgical procedures used to treat bilateral vocal fold motion impairment vary greatly. In a series of 69 patients treated
with endoscopic subtotal arytenoidectomy, PlouinGaudon et al24 reported no problems with voice or
aspiration and reported that all of their patients with
tracheotomy were decannulated. Most other authors
have not reported such a high rate of success. Ossoff
et al4 reported sustained airway restoration in 24 of
28 patients treated with laser arytenoidectomy. Limitations were due to scar contracture and intraluminal granulation.4 In a series of 25 patients with bilateral paralysis treated with cordotomy, Laccourreye
et al25 reported significant airway improvement in 9
of 15 patients who had unilateral cordotomy, and in
10 of 10 patients who had bilateral cordotomy. Of
note, all of the patients in their series were reported
to have severe airway obstruction, but none had a
tracheotomy before surgery.25
In our combined series, AAb was successful in 7
of the 8 patients who did not have a preexisting tracheotomy. However, only 5 of the 9 patients with a
preexisting tracheotomy were decannulated. Factors
noted in patients with poor airway results included
paradoxical vocal fold adduction, prolonged tracheotomy dependence, and poor lung function.
Patients who have had prolonged tracheotomy
have had long-standing vocal fold immobility and
could therefore have fibrotic contracture of adductor muscles. Such contracture would prevent lateralization of the arytenoid cartilage by the abduction
suture, even if the joint is not ankylosed. Paralysis
of long duration has also been associated with poor
outcomes for arytenoid adduction in patients with
unilateral laryngeal paralysis.26
It is not surprising that paradoxical vocal fold adduction would impair results. The AAb suture does
not immobilize the arytenoid cartilage in all dimensions, and active adduction is still possible. Thus,
inspiratory adductor activity can significantly restrict the airway. For example, the patient with adductor breathing dystonia had persistent stridor after
this procedure. A preoperative electromyogram and
careful assessment by flexible endoscopy are recommended to detect possible inspiratory adductor activity so that patients can be appropriately counseled
on the potential outcome. Botulinum toxin alone
has been reported as a treatment for airway obstruction in patients with bilateral laryngeal paralysis by
blocking the activity of adductor muscles.8 In theory, such treatment could improve the results for AAb
surgery. In this series, botulinum toxin was injected
into adductor muscles of 2 of our patients who did
not have sufficient airway improvement after AAb.
Patient 1 had persistent stridor after AAb for bilateral paralysis and did not have improvement after
subsequent endoscopic cordotomy and arytenoidectomy. Botulinum toxin injection was also ineffective. An electromyogram documented strong adductor activity on the left side, and 20 units of toxin
were injected into that side. The patient’s stridor was
not improved, and tracheotomy was performed. It is
possible that she had some contracture of adductor
muscles. Patient 14 was a 70-year-old man with dystonic inspiratory adduction of the larynx. His stridor
persisted after multiple treatments with various doses of botulinum toxin and also after bilateral AAb. It
appeared in the latter case that his stridor persisted
because of contraction of supraglottic muscles.
Arytenoid abduction is clearly more technically
difficult and potentially has more postoperative morbidity than endoscopic cordectomy or arytenoidectomy. Its major advantage is restoration of laryngeal motion by unmasking residual adductor activity.
Theoretically, it should result in a better airway and
a stronger voice than other procedures, so long as
the patient does not have unfavorable laryngeal adductor activity. Furthermore, it should not result in
aspiration, because it does not remove tissue from
the glottis and does not preclude dynamic closure
during the swallow. It provides a stable, adjustable,
and potentially reversible enlargement of the airway. Finally, because there are no intraluminal raw
surfaces or mucosal incisions, there is minimal risk
of intraluminal scarring or granulation tissue.
CONCLUSIONS
Arytenoid abduction is an effective option in the
management of patients with airway obstruction
due to bilateral laryngeal paralysis. It significantly
enlarges the airway without creating an intraluminal mucosal defect, and does not abolish residual
phonatory adduction. It may be less effective in the
presence of active paradoxical vocal fold adduction or prolonged tracheotomy. Further experience
in a larger population could confirm the good voice
outcomes and lack of glottal incompetence during
swallow.
Woodson, Arytenoid Abduction Revisited
37
REFERENCES
1. Jackson C. Ventriculocordectomy: a new operation for
the cure of goitrous paralytic laryngeal stenosis. Arch Surg
1922;4:257-74.
2. King BT. New and function-restoring operation for bilateral abductor cord paralysis: preliminary report. JAMA 1939;
112:814-23.
3. Woodman D. A modification of the extralaryngeal approach to arytenoidectomy for bilateral abductor paralysis. Arch
Otolaryngol 1946;43:63-5.
4. Ossoff RH, Duncavage JA, Shapshay SM, Krespi YP,
Sisson GA Sr. Endoscopic laser arytenoidectomy revisited. Ann
Otol Rhinol Laryngol 1990;99:764-71.
5. Thornell WC. Intralaryngeal approach for arytenoidectomy in bilateral abductor vocal cord paralysis. Trans Am Acad
Ophthalmol Otolaryngol 1949;53:631-6.
6. Dennis DP, Kashima H. Carbon dioxide laser posterior
cordectomy for treatment of bilateral vocal cord paralysis. Ann
Otol Rhinol Laryngol 1989;98:930-4.
7. Lichtenberger G. Reversible lateralization of the paralyzed vocal cord without tracheostomy. Ann Otol Rhinol Laryngol 2002;111:21-6.
8. Ekbom DC, Garrett CG, Yung KC, et al. Botulinum toxin
injections for new-onset bilateral vocal fold motion impairment
in adults. Laryngoscope 2010;120:758-63.
9. Hillel AD, Benninger M, Blitzer A, et al. Evaluation and
management of bilateral vocal cord immobility. Otolaryngol
Head Neck Surg 1999;121:760-5.
10. Eckel HE, Thumfart M, Wassermann K, Vössing M,
Thumfart WF. Cordectomy versus arytenoidectomy in the management of bilateral vocal cord paralysis. Ann Otol Rhinol Laryngol 1994;103:852-7.
11. Worley G, Bajaj Y, Cavalli L, Hartley B. Laser arytenoidectomy in children with bilateral vocal fold immobility. J
Laryngol Otol 2007;121:25-7.
12. Al-Fattah HA, Hamza A, Gaafar A, Tantawy A. Partial
laser arytenoidectomy in the management of bilateral vocal
fold immobility: a modification based on functional anatomical
study of the cricoarytenoid joint. Otolaryngol Head Neck Surg
2006;134:294-301.
13. Tucker HM. Human laryngeal reinnervation: long-term
experience with the nerve-muscle pedicle technique. Laryngo-
scope 1978;88:598-604.
14. Crumley RL. Experiments in laryngeal reinnervation.
Laryngoscope 1982;92(suppl 30):1-27.
15. van Lith-Bijl JT, Stolk RJ, Tonnaer JA, Groenhout C,
Konings PN, Mahieu HF. Laryngeal abductor reinnervation
with a phrenic nerve transfer after a 9-month delay. Arch Otolaryngol Head Neck Surg 1998;124:393-8.
16. Marie JP, Dehesdin D, Ducastelle T, Senant J. Selective
reinnervation of the abductor and adductor muscles of the canine larynx after recurrent nerve paralysis. Ann Otol Rhinol
Laryngol 1989;98:530-6.
17. Faaborg-Andersen K. The position of paretic vocal cords.
Acta Otolaryngol 1964;57:50-4.
18. New GB, Childrey JH. Paralysis of the vocal cords: a
study of 217 medical cases. Arch Otolaryngol 1932;16:143-9.
19. Woodson GE. Configuration of the glottis in laryngeal paralysis. II: Animal experiments. Laryngoscope 1993;103:123541.
20. Woodson GE. Spontaneous laryngeal reinnervation after
recurrent laryngeal or vagus nerve injury. Ann Otol Rhinol Laryngol 2007;116:57-65.
21. Bryant NJ, Woodson GE, Kaufman K, et al. Human posterior cricoarytenoid muscle compartments. Anatomy and mechanics. Arch Otolaryngol Head Neck Surg 1996;122:1331-6.
22. Woodson GE, Picerno R, Yeung D, Hengesteg A. Arytenoid adduction: controlling vertical position. Ann Otol Rhinol
Laryngol 2000;109:360-4.
23. Woodson G, Weiss T. Arytenoid abduction for dynamic
rehabilitation of bilateral laryngeal paralysis. Ann Otol Rhinol
Laryngol 2007;116:483-90.
24. Plouin-Gaudon I, Lawson G, Jamart J, Remacle M. Subtotal carbon dioxide laser arytenoidectomy for the treatment
of bilateral vocal fold immobility: long-term results. Ann Otol
Rhinol Laryngol 2005;114:115-21.
25. Laccourreye O, Paz Escovar MI, Gerhardt J, Hans S,
Biacabe B, Brasnu D. CO2 laser endoscopic posterior partial
transverse cordotomy for bilateral paralysis of the vocal fold.
Laryngoscope 1999;109:415-8.
26. Woodson GE, Murry T. Glottic configuration after arytenoid adduction. Laryngoscope 1994;104:965-9.
Vibratory Asymmetry in Mobile Vocal Folds:
Is It Predictive of Vocal Fold Paresis?
C. Blake Simpson, MD; Linda Seitan May, MD; Jill K. Green, MS;
Robert L. Eller, MD; Carlayne E. Jackson, MD
Objectives: The purpose of this study was to determine whether the videostroboscopic finding of vibratory asymmetry in
mobile vocal folds is a reliable predictor of vocal fold paresis. In addition, the ability of experienced reviewers to predict
the distribution (left/right/bilateral) of the paresis was investigated.
Methods: This is a retrospective chart review of all patients who presented to our clinic during a 3-year period with
symptoms suggestive of glottal insufficiency (vocal fatigue or reduced vocal projection) accompanied by the videostroboscopic findings of bilateral normal vocal fold mobility and vibratory asymmetry. Twenty-three of these patients underwent diagnostic laryngeal electromyography of the thyroarytenoid and cricothyroid muscles to determine the presence
of vocal fold paresis.
Results: Nineteen of the 23 patients (82.6%) were found to have electrophysiological evidence of vocal fold paresis,
either unilaterally or bilaterally, when videostroboscopic asymmetry was present in mobile vocal folds. However, the
three expert reviewers’ ability to predict the distribution (left/right/bilateral) of the paresis was poor (26.3%, 36.8%, and
36.8%, respectively).
Conclusions: The videostroboscopic finding of vibratory asymmetry in mobile vocal folds is a reliable predictor of vocal fold paresis in most cases. However, the ability of expert reviewers to determine the distribution (left/right/bilateral)
of the paresis using videostroboscopic findings is poor. This study highlights the value of laryngeal electromyography in
arriving at a correct diagnosis in this clinical situation.
Key Words: electromyography, videostroboscopy, vocal fold paralysis, vocal fold paresis.
nocent asymmetries [on laryngoscopy] from significant findings may present the greatest challenge in
defining vocal fold paresis.”7(p159) The clinical setting of glottal insufficiency symptoms and grossly
intact vocal fold mobility has previously been described. In these cases, vibratory asymmetry may be
the only laryngoscopic clue to suggest VFP.7 Identification of the asymmetry may help guide the clinician toward performing LEMG and eventually confirming a diagnosis of VFP.
INTRODUCTION
Vocal fold paresis (VFP) is a well-established, albeit controversial, entity. Its incidence is not well
established, but it is likely rare. The few reports that
are available in the literature have shown a range of
as many as 29 cases in a year to as few as 13 cases
over 4 years in tertiary laryngology practices.1-4 Although all of these studies used laryngeal electromyography (LEMG) to confirm the diagnosis, clinicians often use subtle asymmetries on videostroboscopy as indicators that paresis is likely present. During videostroboscopic examination, reduced vocal
fold movement (adduction or abduction), vocal fold
bowing, incomplete glottal closure, and vibratory
asymmetry can all be associated with VFP.4,5 Rubin
et al6 have also described the use of repetitive phonatory tasks to induce fatigue as a means of bringing
out hypomobility in paretic vocal folds. As pointed
out by Sulica and Blitzer, however, “Separating in-
The purpose of this study was to determine whether the videostroboscopic finding of vibratory asymmetry in mobile vocal folds was a reliable predictor of VFP. In addition, the ability of experienced
reviewers to predict the distribution (left/right/bilateral) of the paresis was investigated.
METHODS
Institutional Review Board approval was obtained
From the Departments of Otolaryngology–Head and Neck Surgery (Simpson, May, Green) and Neurology (Jackson), University of
Texas Health Science Center–San Antonio, and the Department of Otolaryngology–Head and Neck Surgery, Wilford Hall Medical Center, Lackland Air Force Base (Eller), San Antonio, Texas.
Presented at the meeting of the American Broncho-Esophagological Association, Las Vegas, Nevada, April 28-29, 2010.
Correspondence: C. Blake Simpson, MD, Dept of Otolaryngology–Head and Neck Surgery, University of Texas Health Science Center, Medical Arts and Research Center, 8300 Floyd Curl Dr, San Antonio, TX 78229.
38
Simpson et al, Vibratory Asymmetry in Vocal Folds
from our institution before the study period. A retrospective chart review was carried out for all patients
who presented to our clinic during a 3-year period
and underwent LEMG for suspected vocal fold paresis.
Over the study period, 48 patients with suspected
VFP underwent diagnostic LEMG. Of those, 23 patients met the study criteria with symptoms of VFP
(vocal fatigue or reduced vocal projection) accompanied by the videostroboscopic findings of bilateral normal vocal fold mobility and vibratory asymmetry. The diagnostic LEMG examinations included an evaluation of the motor unit morphology and
recruitment of motor unit potentials (MUPs) for the
thyroarytenoid and cricothyroid muscles. Interpretation of the LEMG findings was done by a neurologist (C.E.J.) who was blinded to the findings of
the laryngoscopic examination. In all cases, abnormal LEMG findings were considered to be present
when there were large-amplitude polyphasic MUPs
and incomplete recruitment of MUPs. All abnormal
LEMG findings were then classified as left, right,
or bilateral, depending on the side of involvement.
We did not distinguish between recurrent laryngeal nerve (RLN) and superior laryngeal nerve (SLN)
neuropathy for the purposes of this portion of the
study. In other words, if the RLN, SLN, or both
showed electrophysiological evidence of denervation, the findings were considered “abnormal” for
that side.
Our endoscopic clinical examination protocol was
as follows. All of the patients underwent videostroboscopy by means of a flexible laryngoscope with
a distal chip (Olympus ENF-VQ, Olympus Surgical, Orangeburg, New York) rhinolaryngoscope, and
most also had rigid laryngoscopy with a 70° rigid
endoscope (KayPENTAX, Lincoln Park, New Jersey). The patients were instructed to phonate /i/ at
low, modal, and high frequencies. When indicated,
the technique of “unloading” as described by Koufman8 was also used to help reveal more subtle vibratory asymmetry that may have been hidden under
compensatory muscle tension patterns.
When retrospective evaluation of the endoscopic segments was carried out, the following protocol was used. The best-quality videostroboscopic
examination (either flexible or rigid) was used for
each case. Of the 48 cases in which LEMG was performed for suspected paresis, 23 examinations that
were considered to show isolated vibratory asymmetry were selected for the study. The other 25 cases, which showed vocal fold immobility, partial immobility, videostroboscopic evidence of incomplete
closure, or vocal fold lesions, were excluded.
39
TABLE 1. VOCAL FOLD PARESIS DEMOGRAPHICS
AND LEMG FINDINGS
Age
(y)
Gender
Duration
LEMG Findings
Cause of
Paresis
62
67
30
36
28
65
36
69
35
36
44
29
58
37
51
43
76
58
54
F
F
M
M
M
M
F
F
F
M
F
F
F
F
F
F
M
M
F
1y
1y
9y
36 y
4 mo
6y
10 y
2 mo
1y
7y
9y
1.5 y
9 mo
1y
5y
16 mo
6 mo
14 mo
4 mo
B RLN + SLN
B RLN
L RLN + SLN
B RLN
B RLN
B RLN + SLN
B RLN
B RLN
B RLN
B RLN
R RLN
L RLN
L RLN
B RLN
L RLN
R RLN
B RLN
B RLN
L SLN
Idiopathic
Idiopathic
Idiopathic
Congenital
Idiopathic
Idiopathic
Idiopathic
Idiopathic
Idiopathic
Idiopathic
Idiopathic
Idiopathic
Idiopathic
Idiopathic
Idiopathic
Idiopathic
Idiopathic
Idiopathic
Traumatic
LEMG — laryngeal electromyography; B — bilateral; RLN — recurrent laryngeal nerve paresis; SLN — superior laryngeal nerve paresis; L — left; R — right.
The videos were edited to include only segments
in which the vocal folds were in a fully adducted
position and were engaged in vibratory activity. We
decided not to show footage of vocal fold mobility,
in order to help exclude any possible bias that could
occur from interpreting vocal fold movement. The
video segments were then randomized and were interpreted by three reviewers with extensive experience in videostroboscopic interpretation. Each video
segment was reviewed, and the following questions
were addressed: 1) Is asymmetry of vibration (amplitude or mucosal wave) present? 2) If vibration is
asymmetric, which side has the increased amplitude
and/or mucosal wave? and 3) On which side would
you predict the paresis to be present?
The LEMG results were used as the gold standard
for the diagnosis of VFP. Interpretation of the videostroboscopic findings by our reviewers was then
compared to this gold standard to determine the predictive value of subjective vibratory asymmetry on
videostroboscopic examination.
RESULTS
Of the 19 patients with a diagnosis of LEMG-confirmed VFP (Table 1), the mean patient age was 48.5
years (range, 28 to 76 years). Twelve of the patients
were female (63.2%) and had a mean age of 48.8
years, and 7 patients were male (36.8%) and had a
mean age of 47 years. The mean time interval from
40
Simpson et al, Vibratory Asymmetry in Vocal Folds
TABLE 2. LEMG RESULTS AND REVIEWERS’
INTERPRETATION
Patient
1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
Reviewer 1
L
R
R
R
R
R
R
L
L
B
R
L
R
L
R
L
R
R
L
R
R
R
R
Reviewer 2
R
R
R
B
B
R
R
L
R
B
B
L
B
B
B
L
B
L
B
B
L
B
B
Reviewer 3
R
R
R
R
B
R
R
L
B
B
B
L
L
B
R
R
B
L
B
R
R
B
L
LEMG
B
B
L
Normal
B
Normal
B
B
R
B
B
B
R
Normal
L
L
B
Normal
L
R
B
B
L
L — left-sided paresis; R — right-sided paresis; B — bilateral paresis.
the onset of symptoms to presentation to our clinic
was 4.8 years (range, 2 months to 36 years). The
cause of the paresis was idiopathic in the vast majority of cases (17 of 19 or 89.5%), and the remaining
cases were congenital (1 of 19 or 5.2%) or traumatic
(1 of 19 or 5.2%). In terms of neural involvement,
the majority of cases involved the RLN only. Ten
cases were bilateral RLN paresis, and 5 cases were
unilateral RLN paresis. The remaining cases were 2
cases of bilateral combined RLN and SLN paresis, 1
case of unilateral combined RLN and SLN paresis,
and 1 case of unilateral SLN paresis.
Of the 23 patients with symptoms of glottal insufficiency and isolated vibratory asymmetry on videostroboscopy, 19 (82.6%) were found to have electrophysiological evidence of denervation of one or
both vocal folds (Table 2). However, the individual
reviewers’ ability to correctly predict the distribution
of the paresis was quite poor. Given three options
(bilateral, left, or right), each reviewer was unable
to correctly predict the side in most cases (reviewer
1, 5 of 19 correct; reviewer 2, 7 of 19 correct; and
reviewer 3, 7 of 19 correct). With all examination
evaluations combined, the side of paresis was correctly predicted in only 33.3% of cases (19 of 57).
DISCUSSION
The idea behind this study was to answer a com-
mon question that is posed in our multidisciplinary
clinics. As a general rule, the voice team (which includes the senior author, speech pathologist, and resident physician) reviews the videostroboscopic examination of the patient and discusses the subjective
interpretation of the vibratory parameters. In most
cases of suspected VFP, the clinicians can agree that
vibratory asymmetry is present, and LEMG will
later confirm the diagnosis. However, the reliability of using vibratory asymmetry to correctly predict the presence of VFP has not been examined.
Although we can usually agree on the presence of
vibratory asymmetry, there is often a debate about
the sidedness of the suspected paresis. Conventional
thinking suggests that the denervated side will have
an increased amplitude and/or mucosal wave due to
the laxity of the paretic vocal fold. Despite this consensus, we have noted that many times the clinicians
do not agree as to which side(s) is involved.
Obviously, the clinical diagnosis of some cases of
VFP is fairly straightforward when based on videostroboscopic findings and clinical history. In the setting of gross hypomobility and glottal insufficiency, the diagnosis is not often in question. However,
when there are no readily apparent differences in vocal fold mobility, the diagnosis can be more difficult
to make, or may not be suspected by the clinician
at all. In these cases, vibratory asymmetry may be
the only clue that VFP is present.7 This finding may
help guide the clinician toward performing LEMG
and establishing a correct diagnosis.
Our clinical protocol for patients with symptoms
suggestive of glottal insufficiency and an increased
amplitude and/or mucosal wave or “chasing wave”
(asymmetry of vibration) is to recommend LEMG.
Obviously, not all patients with this combination of
symptoms and findings agree to undergo or follow
up for diagnostic LEMG, so we are not able to comment on the positive predictive value of vibratory
asymmetry in these cases. Nonetheless, when vibratory asymmetry prompted LEMG testing in our series, the clinical “hunch” ended up being correct in
83% of cases. However, the ability of experienced
clinicians to correctly predict which side was involved was quite poor (33.3%). This is exactly the
percentage one would expect if the clinician’s determination were randomly generated; ie, there is a
1-in-3 chance of predicting the outcome correctly.
The difficulty partially arises from using the subjective observation that one side demonstrates increased vibratory amplitude (often thought to be a
manifestation of reduced muscular tone in a denervated vocal fold). By necessity, that determination
involves using the contralateral side as a control,
ie, the side with the “normal tone.” In many cas-
Simpson et al, Vibratory Asymmetry in Vocal Folds
es, however, this side may also be affected, making the assumption of unilaterality erroneous. Despite this problem, there were many cases in which
the reviewer correctly predicted that the paresis was
unilateral, but the predicted side (ie, distribution of
involvement) was incorrect.
Relying solely on laryngoscopic findings to predict VFP continues to be problematic. Other studies
have shown that 25% to 40% of patients had LEMG
findings that were not predicted by their laryngoscopic examination.2,3 Although vibratory asymmetry is fairly predictive of VFP (83% of cases in our
study), determining the distribution (left/right/bilateral) of the paresis is very poorly predictive.
Interpretation of videostroboscopic examinations
is by nature subjective. We have observed that vibratory asymmetry can sometimes be difficult to detect on routine stroboscopy. The best method of accentuating asymmetry is to have the patient phonate
41
at a modal or low fundamental frequency at a high
intensity. In addition, extinguishing any secondary
supraglottic muscular tension seems to be beneficial, as this allows for the differential tension of the
true vocal folds to be observed. Last, recording the
examination and playing it back in slow motion, or
performing frame-by-frame analysis, is yet another
method to aid in the detection of vibratory asymmetry.
CONCLUSIONS
The videostroboscopic finding of vibratory asymmetry in mobile vocal folds is a reliable predictor of
VFP in most cases. However, the ability of expert
reviewers to determine the distribution (left/right/
bilateral) of the paresis using videostroboscopic
findings is poor. This finding highlights the value
of LEMG in arriving at a correct diagnosis in this
clinical situation.
REFERENCES
1. Merati AL, Shemirami N, Smith TL, Toohill RJ. Changing trends in the nature of vocal fold motion impairment. Am J
Otolaryngol 2006;27:106-8.
2. Heman-Ackah YD, Barr A. Mild vocal fold paresis: understanding clinical presentation and electromyographic findings. J Voice 2006;20:269-81.
3. Koufman JA, Postma GN, Cummins MM, Blalock DP.
Vocal fold paresis. Otolaryngol Head Neck Surg 2000;122:53741.
4. Simpson CB, Cheung EJ, Jackson CJ. Vocal fold paresis:
clinical and electrophysiologic features in a tertiary laryngology
practice. J Voice 2009;23:396-8.
5. Altman KW. Laryngeal asymmetry on indirect laryngos-
copy in a symptomatic patient should be evaluated with electromyography. Arch Otolaryngol Head Neck Surg 2005;131:35660.
6. Rubin AD, Praneetvatakul V, Heman-Ackah Y, Moyer
CA, Mandel S, Sataloff RT. Repetitive phonatory tasks for identifying vocal fold paresis. J Voice 2005;19:679-86.
7. Sulica L, Blitzer A. Vocal fold paresis: evidence and controversies. Curr Opin Otolaryngol Head Neck Surgery 2007;15:
159-62.
8. Koufman JA. Evaluation of laryngeal biomechanics by fiberoptic laryngoscopy. In: Rubin JA, Sataloff RT, Korovin GS,
Gould WJ, eds. Diagnosis and treatment of voice disorders.
New York, NY: Igaku-Shoin, 1995:122-34.
Effects of Nerve-Muscle Pedicle on Immobile Rat Vocal Folds
in the Presence of Partial Innervation
Takashi Aoyama, MD; Yoshihiko Kumai, MD, PhD; Eiji Yumoto, MD, PhD;
Takaaki Ito, MD, PhD; Satoru Miyamaru, MD, PhD
Objectives: We investigated whether implantation of an ansa cervicalis nerve (ACN)–muscle pedicle into the thyroarytenoid (TA) muscle is efficacious in the presence of partial recurrent laryngeal nerve (RLN) innervation.
Methods: We studied a total of 36 rats. Twelve of the rats served as positive and negative control animals. In the remaining 24 rats, the left RLN was transected, a 1-mm piece of nerve was removed, and the stumps were abutted in silicone
tubes (STs), inducing partial RLN regeneration. Twelve of the ST-treated rats underwent this procedure alone, and the
other 12 rats had a nerve-muscle pedicle (NMP) implanted into the left TA muscle 5 weeks after ST treatment. At 15
weeks, reinnervation was assessed by histologic evaluation of the TA muscle and by electromyography with stimulation
of the RLNs and ACNs.
Results: The muscle area, the number of nerve terminals, the number of acetylcholine receptors, and the ratio of nerve
terminals to acetylcholine receptors were significantly greater (p < 0.05) in the NMP group than in the ST group. Electromyography elicited TA muscle compound action potentials upon stimulation of the RLNs and ACNs.
Conclusions: In rats, NMP implantation is efficacious for reducing atrophic changes in the TA muscle in the presence of
partial RLN innervation.
Key Words: nerve-muscle pedicle, neuromuscular junction, partial denervation, recurrent laryngeal nerve paralysis.
on a partially innervated TA muscle is questionable
because of synkinetic RLN reinnervation, which is
common clinically. Therefore, it is necessary to determine whether an NMP can innervate a TA muscle
that is partially innervated by the RLN.
INTRODUCTION
Unlike the complete loss of recurrent laryngeal
nerve (RLN) innervation, which leads to marked
thyroarytenoid (TA) muscle atrophy, many RLN injuries cause partial denervation and reinnervation
with various degrees of vocal fold motion impairment, accompanied by weakness, synkinesis, and
partial atrophy.1 Successful clinical application of
the nerve-muscle pedicle (NMP) implantation method for patients with unilateral vocal fold paralysis
has been reported.2 Previously, we demonstrated
that ansa cervicalis nerve (ACN) NMP flap implantation into the TA muscle prevented the denervation
and atrophy associated with complete RLN denervation in rats.3,4 Therefore, the NMP procedure can
enable effective motor innervation following complete denervation. In other nonlaryngeal muscles,
implanted foreign nerves will partially reinnervate
denervated muscles.5
MATERIALS AND METHODS
Animal Preparation and Endoscopic Observation. Thirty-six 8-week-old female Wistar rats were
used. The animals were housed at a constant temperature (22°C) on a 12-hour light-dark cycle and were
given food and water as desired. All experiments
were conducted in accordance with the guidelines
of the Animal Care and Use Committee in compliance with the Animal Research Center at Kumamoto University’s Graduate School of Medicine, Kumamoto, Japan. First, the vocal folds of the animals
were examined endoscopically (Nisco, Tokyo, Japan) to check for spontaneous motion of the bilateral vocal folds while anesthetized intraperitoneally
with ketamine hydrochloride (50 mg/kg) and xylazine hydrochloride (10 mg/kg). Six animals without
left RLN transection served as a control group. The
Nevertheless, it is possible that innervated, functionally active muscle fibers will not accept further
innervation.6 If so, the effect of the NMP procedure
From the Departments of Otolaryngology–Head and Neck Surgery (Aoyama, Kumai, Yumoto, Miyamaru) and Pathology and Experimental Medicine (Ito), Kumamoto University School of Medicine, Kumamoto, Japan.
Presented at the meeting of the American Broncho-Esophagological Association, Las Vegas, Nevada, April 28-29, 2010.
Correspondence: Yoshihiko Kumai, MD, PhD, Dept of Otolaryngology–Head and Neck Surgery, Kumamoto University School of
Medicine, 1-1-1 Honjo, Kumamoto City, Kumamoto 860-8556, Japan.
42
Aoyama et al, Nerve-Muscle Pedicle in Partially Innervated Larynx
A
43
Fig 1. A) Silicone tube (ST) model. One millimeter of left recurrent laryngeal nerve (RLN)
was resected at level of seventh tracheal ring,
and both nerve stumps were passed into ST.
Photograph is magnified view of ST. B) Nervemuscle pedicle (NMP) model. Five weeks after
ST implantation, NMP flap was implanted in
exposed thyroarytenoid (TA) muscle through
window made in thyroid cartilage. Photograph
shows rat larynx after ST procedure and NMP
implantation.
B
other 30 animals were anesthetized as described
above. The strap muscles were exposed via a vertical midline skin incision. Under an operating microscope (SN-MD2; Nagashima Medical Instruments,
Tokyo), the sternohyoid muscles were divided at
the midline. After exposure of the left RLN, a 1-mm
length was resected at the level of the seventh tracheal ring. In 6 of the transected animals (complete
denervation group), the distal end of the nerve was
ligated with 4-0 nylon suture, whereas the proximal
end was embedded in the sternohyoid muscle to
prevent contact between the cut ends, as described
previously.3 In the remaining 24 animals, both nerve
stumps were placed in a silicone tube (ST) and fixed
to it with 9-0 nylon suture. After the procedure, left
vocal fold immobility was confirmed endoscopically and the skin incision was closed. The animals,
which were allowed to recover in an approved animal care facility, were divided into 2 groups: 12 that
underwent anastomosis only, with STs (ST group;
Fig 1A), and 12 in which, under an operating microscope, the ACN branch was harvested with a 0.5 ×
0.5 × 0.5-mm piece of sternohyoid muscle and implanted in the TA muscle through a small window
made in the thyroid cartilage. An NMP flap was
firmly sutured to the surface of the TA muscle by
use of 9-0 nylon suture, as previously described,2,3
5 weeks after anastomosis (NMP group; Fig 1B).
Each group was divided into 2 groups of 6, one for
histologic analysis and the other for evoked electromyography (EMG).
Evoked EMG. At 15 weeks after the ST procedure, reinnervation was assessed in the NMP group
by EMG with stimulation of the right RLN, left
RLN, and ACN, as described elsewhere,4,7 before
and after transection of the left RLN and ACN (Fig
2). A monopolar platinum electrode, 0.08 mm in diameter and coated with silicone, was inserted into
the TA muscle through the thyroid cartilage. Evoked
EMG signals were recorded through a Neuropack
(MEB-9102; Nihon Kohden, Tokyo) combined with
an electric stimulator (SEN-3301; Nihon Kohden).
A single 100-μs pulse of supramaximal intensity
was applied for stimulation.
Area of TA Muscle and Individual Muscle Fibers.
We next examined the areas of the entire TA muscle
and individual muscle fibers in the total denervation,
ST, and NMP groups, as earlier described.3,4 The areas were determined on the basis of pixel numbers
44
A
Aoyama et al, Nerve-Muscle Pedicle in Partially Innervated Larynx
B
Fig 2. A) Electrically stimulated (point α — treated RLN; point β — ansa cervicalis nerve [ACN]; point γ — untreated RLN)
and transected (point a — treated RLN; point b — ACN) portions were subjected to evoked electromyography. B) Representative evoked electromyographic findings for left and right TA muscles in NMP group at 15 weeks. 1) Waveform with stimulation
of point α before RLN transection. 2) Waveform with stimulation of point α after RLN transection at point a. 3) Waveform with
stimulation of point β before ACN transection. 4) Waveform with stimulation of point β after ACN transection at point b. 5)
Waveform with stimulation of point γ of RLN on untreated side.
by tracing the outlines of the TA muscle and its individual fibers with the Image software program
(National Institutes of Health, Bethesda, Maryland).
Atrophic changes in the TA muscle were evaluated
by comparing the areas from the treated (left, T) and
untreated (right, U) sides (T/U ratio). In the control
group, the areas of the complete TA muscle and its
individual muscle fibers were evaluated by comparing both sides as described above.
In addition, the percentage of muscle fibers containing centrally located nuclei (based on 100 muscle fibers from a single animal) was determined for
the treated side in all animals, and the results for
each group were averaged.
Neuromuscular Junctions. The numbers of neuromuscular junctions (NMJs), nerve terminals (NTs),
and acetylcholine receptors (AchRs) in the groups
were determined as earlier described.3,4 Sections
were incubated with polyclonal rabbit anti-human
synaptophysin antibodies (Dako, Tokyo) to label
NTs, and with rhodamine-conjugated α-bungarotoxin (Sigma, Tokyo) to label AchRs. We calculated
the T/U ratios for the NTs and AchRs and the NT/
AchR ratio on the treated side in each group.
Statistical Analysis. All data were subjected to
unpaired Student’s t-tests and are presented as the
mean ± SD. Differences were considered statistical-
ly significant at p < 0.05.
RESULTS
Endoscopic Observation. Fifteen weeks after
RLN transection, no spontaneous vocal fold movement was noted on the left side in any of the animals
in the denervation, ST, and NMP groups.
Evoked EMG. In the NMP group, when the RLN
proximal to the ST was stimulated before its transection, evoked action potentials were seen in the TA
muscle (Fig 2B, row 1), whereas no muscle activity
was elicited when the same portion of the RLN was
stimulated after the RLN was transected at point “a”
(Fig 2B, row 2). Conversely, when the transplanted
ACN was stimulated at point “β,” evoked action potentials were seen in the TA muscle (Fig 2B, row
3); however, no muscle activity was elicited when
the distal transected ACN was stimulated at the
same portion (Fig 2B, row 4). On the untreated side,
evoked action potentials were seen in the TA muscle
(Fig 2B, row 5).
Areas of TA Muscle and Individual Muscle Fibers. In the NMP group, the total TA muscle area
on the treated side (Fig 3D) was smaller than that in
the control animals (Fig 3A), but larger than those
in both the denervation and ST groups (Fig 3B,C).
Magnified images of the TA muscle revealed that
Aoyama et al, Nerve-Muscle Pedicle in Partially Innervated Larynx
45
A
B
C
D
E
F
G
H
Fig 3. Representative microscopic findings of TA muscle with hematoxylin and eosin staining. A,E) Control group. B,F) Group
at 15 weeks with complete RLN denervation procedure alone. C,G) ST group at 15 weeks with ST procedure alone. D,H) NMP
group at 15 weeks with ST procedure and NMP implantation at 5 weeks. Ultimately, evaluation times were same (23-week-old
animals). Scale bars — 200 μm in A-D; 20 μm in E-H.
the areas of individual muscle fibers on the treated
side (Fig 3H) were similar to those in the control
group (Fig 3E), and larger than those in the denervation and ST groups (Fig 3F,G). The T/U ratio for
the entire TA muscle area in the NMP group was
significantly greater than that in the ST group (p <
0.01; Fig 4A). Although the difference did not reach
statistical significance, the T/U ratio for the areas
of individual muscle fibers in the NMP group was
slightly larger than that in the ST group (p = 0.051;
Fig 4B).
Centralized Nuclei in Muscle Fibers. The incidence of centralized nuclei in the fibers on the treated side in the NMP group was significantly greater
than those in the control, denervation, and ST groups
(p < 0.01; Fig 5).
Neuromuscular Junctions. A number of synaptophysin-positive NTs (Fig 6A-D) and α-bungarotoxin–positive AchRs (Fig 6E-H) were observed in all
groups. The T/U ratios of the number of NTs and
AchRs, and the NT/AchR ratio for the NMP group
(for the quantitative measurement of regenerated
NMJs8), were significantly higher than those for the
denervation and ST groups (p < 0.01; Fig 7).
DISCUSSION
Previously, we demonstrated that ACN NMP implantation into the TA muscle prevented the denervation atrophy associated with complete RLN den-
Fig 4. Comparison of treated-untreated
ratios of TA muscle area for A) entire
muscle and B) individual muscle fibers
between all groups (control, RLN denervation [RD], ST, and NMP groups) at 15
weeks. All data are presented as mean ±
SD. Differences were considered statistically significant at p < 0.05.
A
B
46
Aoyama et al, Nerve-Muscle Pedicle in Partially Innervated Larynx
time period. We assume that decreasing or increasing this time period might bring about a relatively
low degree of variance in NMJ regeneration.
Fig 5. Percentage of muscle fibers containing centrally
located nuclei at 15 weeks, based on 100 fibers in TA
muscle for all groups (control, RD, ST, and NMP groups).
All data are presented as mean ± SD.
ervation in rats.3,4 However, as was shown previously, innervated, functionally active muscle fibers may
not accept further innervation.6 Thus, the effect of
the NMP procedure on a TA muscle that is partially
innervated because of RLN synkinetic reinnervation
is questionable. This prompted us to investigate the
effects of dual innervation, which imply additional
effects of the foreign nerve (ACN NMP) on the reinnervation of a TA muscle partially innervated by the
RLN. We examined the effect of ACN NMP implantation 15 weeks after the ST procedure, because our
previous studies3,4 demonstrated that the prominent
effect of the NMJ regeneration was obtained by this
Injury to the RLN may cause various degrees of
vocal fold motion impairment secondary to ineffective, synkinetic muscle activity, rather than chronic
denervation or marked muscle atrophy1 — a finding
suggesting that partial, misdirected reinnervation of
the intrinsic laryngeal muscles has occurred. Nonfunctional laryngeal reinnervation following RLN
injury, presenting as a completely immobile vocal
fold, may be due to the loss of adductor and abductor fiber organization during regenerative growth
of the RLN.9 Various experimental animal models
have been used to study peripheral nerve injury and
functional recovery with synkinetic muscle activity,10,11 including an animal model of RLN crush
injury using aneurysm clips to simulate synkinetic
muscle activity following intraoperative RLN injury.12 Tessema et al12 described this crush injury
model as a valuable RLN synkinetic reinnervation
model; however, in our preliminary study, endoscopy revealed the recovery of vocal fold mobility
6 or 7 weeks after the procedure (data not shown).
This contradiction (a persistently immobile vocal
fold following RLN injury) was the basis for our
use of the ST model as an RLN synkinetic reinnervation model with an immobile vocal fold. In our
preliminary study, endoscopy revealed consistent
vocal fold immobility, whereas histologic examina-
A
B
C
D
E
F
G
H
Fig 6. Representative microscopic findings of neuromuscular junctions in TA muscle. A,E) Control group. B,F) Denervation
group at 15 weeks with complete RLN denervation alone. C,G) ST group at 15 weeks with ST procedure alone. D,H) NMP
group at 15 weeks with ST treatment and NMP implantation at 5 weeks. Neuromuscular junctions were double-stained with
fluorescein isothiocyanate–labeled synaptophysin (A-D) and rhodamine-labeled α-bungarotoxin (E-H). Scale bars — 20 μm.
Aoyama et al, Nerve-Muscle Pedicle in Partially Innervated Larynx
A
B
47
C
Fig 7. A,B) Temporal changes in treated-untreated ratios of number of A) nerve terminals and B) acetylcholine receptors among
all groups. C) Comparison of ratio of number of nerve terminals to that of acetylcholine receptors on treated side of TA muscle
between all groups (control, RD, ST, and NMP groups). Data are presented as mean ± SD. Differences were considered statistically significant at p < 0.05.
tion showed nerve regeneration through the ST preventing TA muscle atrophy; these data, in addition
to our electrophysiological findings, including the
prolonged latency of the evoked EMG (Fig 2B, row
1) and the reappearance of NMJs, validate this as
an RLN synkinetic reinnervation model with an immobile vocal fold (details to be presented in another
article). We implanted the ACN NMP into the TA
muscle 5 weeks after the ST procedure because of
observations that demonstrate RLN synkinetic reinnervation at this very time point. However, further
investigation is necessary to see whether this waiting period until implantation might be critical to the
efficacy of this procedure, because a longer waiting
period might bring about a lower degree of plasticity
of the NMJ arrangement.
In this study, using the ST model, we found that
NMP implantation is efficacious (in comparison
with the ST group) for reducing atrophic changes
(represented by the muscle area) and increasing synapse formation (represented by the ratio of NTs to
AchRs in the TA muscle), even in the presence of
partial RLN innervation. In addition, EMG showed
that TA compound action potentials could be elicited by stimulating the proximal sides of both the
transected RLN and the transferred ACN. Thus, partially innervated TA muscle fibers can accept foreign motor nerves (ACN) in addition to reinnervation from the original RLN.
A previous study demonstrated that newly fused
satellite cell nuclei were initially centralized in regenerated myofibrils and later migrated to a more
peripheral location.13 Although it is difficult to demonstrate a direct correlation between the presence of
centralized muscle fiber nuclei and the regenerative
capacity of a partially innervated TA muscle by use
of our study design, we postulate that dual neural input into the partially innervated TA muscle occurred
as a result of NMP implantation, on the basis of a
comparison between the numbers of centralized nuclei in the ST and NMP groups (p < 0.01).
It remains to be addressed to what extent reinnervation occurs by sprouting of the foreign nerve
(ACN) into the partially innervated TA muscle. We
postulate that the transplanted ACN sprouts so that
it targets original or newly formed AchRs to form
new NMJs.3,4 Axonal sprouting is the process by
which fine nerve processes grow out from intact
axons at NTs to give rise to terminal sprouts. This
commonly occurs as a natural compensatory mechanism following motor neuron diseases or trauma,
such as partial nerve injuries.14 Previous studies
showed that several factors influence this process,
including the amount of scar tissue that forms at the
implantation neuroma, the number of motor axons
in the donor nerve, and the level of neuromuscular
activity in the recipient muscle.15 It was previously shown that the efficacy of the NMP method for
a partially denervated muscle was critically dependent on the level of neuromuscular activity in the recipient muscle.16 Interestingly, excessive neuromuscular activity significantly reduces the bridging of
perisynaptic Schwann cell processes between innervated and denervated end plates, thereby inhibiting
axonal sprouting in partially denervated muscle.14
By contrast, moderate and chronic increases in muscle activity, such as synkinetic nerve regeneration
or moderate exercise, promote axonal sprouting of
the implanted nerve, which may facilitate functional recovery in partially innervated muscle.16 Thus, a
partially innervated TA muscle due to RLN synkinetic reinnervation, which might show a long-term
48
Aoyama et al, Nerve-Muscle Pedicle in Partially Innervated Larynx
increase in muscle activity, could promote sprouting
from the transplanted ACN, which in turn would increase the NT/AchR ratio with positive EMG findings as a consequence of the newly formed NMJs.
Further investigation is necessary to see whether the
waiting period for the implantation might be critical
for the efficacy of this procedure, because a longer
waiting period might cause a lower degree of plasticity of the NMJ arrangement.
CONCLUSIONS
Histologic and electrophysiological analyses confirmed that reinnervation using the NMP procedure
can reduce atrophic changes in the TA muscle, even
in the presence of partial RLN innervation 5 weeks
after the ST procedure in rats. Thus, the NMP method may be useful for treating patients in whom partial innervation of the TA muscle is suspected.
REFERENCES
1. Crumley R. Laryngeal synkinesis revisited. Ann Otol
Rhinol Laryngol 2000;109:365-71.
2. Tucker HM. Long-term results of nerve-muscle pedicle
reinnervation for laryngeal paralysis. Ann Otol Rhinol Laryngol
1989;98:674-6.
3. Kumai Y, Ito T, Udaka N, Yumoto E. Effects of a nervemuscle pedicle on the denervated rat thyroarytenoid muscle.
Laryngoscope 2006;116:1027-32.
4. Miyamaru S, Kumai Y, Ito T, Sanuki T, Yumoto E. Nervemuscle pedicle implantation facilitates re-innervation of longterm denervated thyroarytenoid muscle in rats. Acta Otolaryngol 2009;129:1486-92.
5. Brown MC, Ironton R. Sprouting and regression of neuromuscular synapses in partially denervated mammalian muscles. J Physiol 1978;278:325-48.
6. Mark RF. Selective innervation of muscle. Br Med Bull
1974;30:122-6.
7. Zealear D, Swelstad M, Fortune S, et al. Evoked electromyographic technique for quantitative assessment of the innervation status of laryngeal muscles. Ann Otol Rhinol Laryngol
2005;114:563-72.
8. Deschenes MR, Will KM, Booth FW, Gordon SE. Unlike
myofibers, neuromuscular junctions remain stable during prolonged muscle unloading. J Neurol Sci 2003;210:5-10.
9. Gacek RR. Morphologic correlates for laryngeal reinner-
vation. Laryngoscope 2001;111:1871-7.
10. Mu L, Yang S. An experimental study on the laryngeal
electromyography and visual observation in varying types of
surgical injuries to the unilateral recurrent laryngeal nerve in the
neck. Laryngoscope 1991;101:699-708.
11. Flint PW, Downs DH, Coltrera MD. Laryngeal synkinesis following reinnervation in the rat. Neuroanatomic and physiologic study using retrograde fluorescent tracers and electromyography. Ann Otol Rhinol Laryngol 1991;100:797-806.
12. Tessema B, Roark RM, Pitman MJ, Weissbrod P, Sharma
S, Schaefer SD. Observations of recurrent laryngeal nerve injury
and recovery using a rat model. Laryngoscope 2009;119:164451.
13. Hawke TJ, Garry DJ. Myogenic satellite cells: physiology to molecular biology. J Appl Physiol 2001;91:534-51. [Erratum in J Appl Physiol 2001;91:2414.]
14. Tam SL, Gordon T. Neuromuscular activity impairs axonal sprouting in partially denervated muscles by inhibiting
bridge formation of perisynaptic Schwann cells. J Neurobiol
2003;57:221-34.
15. Sorbie C, Porter TL. Reinnervation of paralysed muscles
by direct motor nerve implantation. An experimental study in
the dog. J Bone Joint Surg Br 1969;51:156-64.
16. Tam SL, Gordon T. Mechanisms controlling axonal
sprouting at the neuromuscular junction. J Neurocytol 2003;32:
961-74.
Use of a Microdebrider for Subepithelial Excision of
Benign Vocal Fold Lesions
Alex Y. Cheng, MD; Ahmed M. S. Soliman, MD
Objectives: We present a series of patients who underwent excision of subepithelial lesions using a laryngeal microdebrider.
Methods: A retrospective review was carried out of all patients who between June 2005 and April 2009 underwent microlaryngoscopy and excision of vocal fold lesions by the senior author using a microdebrider. The patients’ demographics, past medical history, preoperative and postoperative videostroboscopy findings, and surgical pathology findings were
reviewed.
Results: Eight patients were identified. All were female and had a mean age of 44 years (range, 21 to 54 years). Four
patients had a unilateral pseudocyst, 2 patients had bilateral pseudocysts, 1 patient had bilateral Reinke’s edema, and 1
patient had unilateral Reinke’s edema. Through an incision made just lateral to the lesion on the superior surface, a small
microdebrider blade was inserted and run at 800 to 1,000 revolutions per minute in oscillation mode with low wall suction
until the lesion was completely excised, and the overlying epithelium was left intact. Postoperative videostroboscopic
examination of the larynx revealed an absence of lesions and near-complete glottal closure in all patients. Seven of the 8
patients reported an improvement in voice.
Conclusions: Microdebrider excision of subepithelial vocal fold lesions is feasible. The patients with Reinke’s edema
had slightly more residual edema, a decreased epithelial wave, and less improvement in voice as compared to the patients
with pseudocysts. Meticulous technique, low oscillation speeds, and low wall suction are necessary to avoid injury to the
overlying epithelium.
Key Words: benign lesion, microdebrider, surgical treatment, vocal fold lesion.
Its use in the treatment of Reinke’s edema was first
described in 2000,7 and very recently, functional
outcomes were reported.8
INTRODUCTION
The microdebrider is a powered microdissecting instrument that was originally designed for arthroscopic knee surgeries. With a small oscillating
blade within a protected sheath that provides continuous suction, the microdebrider is able to remove
tissues within a tight space and allow an unobstructed operative field. Further technological advances in
microdebrider design have since allowed it to access
the narrow confines of the upper aerodigestive tract
and thus allowed its use in otolaryngology.
Microdebrider use in endoscopic sinus surgery
was first reported in 1996,1 and it was subsequently
adapted for laryngeal applications in 1999.2 Its first
use in the larynx was as an effective debulking tool
for laryngeal papillomas and later for various other
laryngeal disorders.3-5 More delicate laryngeal surgery using smaller blades at low rotational speeds
has been briefly mentioned in the literature for epithelial lesions, including polyps and granulomas.6
Benign subepithelial disorders of the true vocal
folds include Reinke’s edema, cysts, pseudocysts,
and fibrous masses. Although traditional surgical excision of true cysts using microsurgical techniques is more straightforward, it is more challenging for pseudocysts, of which the wall is thinner and
often adherent to the subepithelium of the elevated
microflap. In Reinke’s edema, the thick gelatinous
matrix is often too thick to remove with suctioning
alone. In these selected situations, we have used the
microdebrider to excise the lesions subepithelially.
We review our experience and technique with the
microdebrider in patients with benign subepithelial
vocal fold lesions.
METHODS
After obtaining Institutional Review Board ap-
From the Department of Otolaryngology–Head and Neck Surgery, Temple University School of Medicine, Philadelphia, Pennsylvania.
Presented at the meeting of the American Broncho-Esophagological Association, Las Vegas, Nevada, April 28-29, 2010.
Correspondence: Ahmed M. S. Soliman, MD, Dept of Otolaryngology–Head and Neck Surgery, Temple University School of Medicine, 3400 N Broad St, Kresge West 102, Philadelphia, PA 19140.
49
50
Cheng & Soliman, Subepithelial Microdebrider Excision
(Patient 2) Endoscopic views of endoscopic subepithelial
shaver excision of pseudocyst. A) Subepithelial lesion is
seen on right true vocal fold. B) After incision alongside lesion with sickle knife, microdebrider is introduced into subepithelial space and lesion is excised. C) Epithelium has
been redraped, leaving smooth edge.
A
B
proval, we carried out a retrospective review of all
patients who underwent microlaryngoscopy and
excision of vocal fold lesions by the senior author
(A.M.S.S.) using the microdebrider from June 2005
to April 2009. The patients’ demographics, past
medical history, and preoperative and postoperative
laryngoscopy findings were reviewed.
After the airway was secured, an operative laryngoscope was inserted and the larynx was suspended
with the true vocal folds in view. Under microscopic
visualization, a sickle knife was used to incise the
epithelium just lateral to the lesion on the superior surface of the vocal fold. An angled blunt elevator was then used to dissect the lesion away from
the underlying superficial lamina propria and vocal
ligament and the overlying epithelium. In cases in
which the lesion was tightly adherent to the overlying epithelium, the Xomed XPS Shaver system with
a Magnum handpiece (Medtronic-Xomed, Jacksonville, Florida) was used. An angled 2.9- or 3.5-mm
round window microdebrider blade (Skimmer) was
inserted and then run at 800 to 1,000 revolutions per
minute in oscillation mode with low wall suction until the lesion was completely excised, and the over-
C
lying epithelium was left intact (see Figure). Strict
care was also taken to keep the underlying superficial lamina propria and vocal ligament undisturbed.
The 3.5-mm blade was used initially until the availability of the 2.9-mm blade, which is now used exclusively. The 2 patients with Reinke’s edema had
the excess epithelium excised sharply with an upbiting microscissors after the gelatinous matrix was
removed with the microdebrider subepithelially.
Rigid or flexible videostroboscopic examination of
the larynx (KayPENTAX, Lincoln Park, New Jersey) was performed before and after the operation
in all patients.
RESULTS
Twelve patients who underwent excision of vocal fold lesions using a microdebrider were identified. Four patients with airway-obstructing polyps
were excluded from the study because these lesions
were not excised subepithelially. The remaining 8
patients were all female and had a mean age of 44
years (range, 21 to 54 years). All patients presented
with a chief complaint of hoarseness. Other symptoms included globus sensation, excessive throat
Cheng & Soliman, Subepithelial Microdebrider Excision
clearing, and inability to sing as usual. Three patients (38%) had a history of smoking.
All patients underwent preoperative videostroboscopy, which revealed a unilateral pseudocyst in
4 patients, bilateral pseudocysts in 2 patients, bilateral Reinke’s edema in 1 patient, and unilateral
Reinke’s edema in 1 patient. All patients were empirically treated with daily proton pump inhibitors
started at their preoperative visit and continued after
the operation.
All patients underwent elective outpatient sameday surgery. They were intubated with a 6.0 endotracheal tube and underwent general anesthesia. Surgical excision was carried out as described in the
surgical technique. The postoperative diagnoses
concurred with the preoperative impression in all
cases.
The first postoperative appointment was at a
mean of 16.3 days from the date of surgery. One patient did not return to the office; however, on telephone interview 2 years after her procedure, she reported her voice to be normal. Of the 7 patients who
returned to the office, 6 (85.7%) reported subjective
improvement in symptoms. Videostroboscopic examination of the larynx revealed an absence of lesions and near-complete glottal closure in all patients. All had a slightly decreased mucosal wave,
as well as mild edema and/or erythema of the operated vocal fold. This finding was more pronounced
in the 2 patients with Reinke’s edema. Postoperative
videostroboscopy of the patient who reported no improvement in voice showed similar findings.
Six patients had a second follow-up office visit (mean, 78.5 days). The patient who reported no
early postoperative improvement continued to have
similar complaints, although her videostroboscopic examination findings were normal; it was later
learned that she was applying for disability benefits
from her job as a telephone operator. Three patients
who had pseudocysts removed reported near-normal
voice quality; their videostroboscopic examinations
revealed a symmetric, periodic mucosal wave with
complete glottal closure and an absence of lesions.
Two patients who had Reinke’s edema reported
marked improvement in voice, but on videostroboscopic examination had mild edema of the affected
vocal folds.
DISCUSSION
The microdebrider has been successfully used in
the larynx, primarily for the debulking of various
lesions, including papillomas, polyps, and granulo-
51
mas. However, its use for excision of subepithelial lesions other than Reinke’s edema has not been
previously reported. We report its application to selected cases as a targeted approach for a surgically challenging situation. Whenever possible, every
attempt was made to remove the lesions by use of
traditional micro-instruments; however, when there
was tight adherence to the overlying epithelium or a
large amount of gelatinous matrix that could not be
suctioned, the microdebrider provided a significant
advantage. It is also important to emphasize that as
in traditional cold microsurgical techniques, every
effort is made to avoid injury to the underlying superficial lamina propria and vocal ligament, which
are most important in vibratory function, as well
as to avoid injury to the overlying epithelium. The
use of adequate magnification and low wall suction
along with the low variable oscillation is critical in
achieving this goal.
Unlike the original descriptions by Sant’Anna
and Mauri7 of its use in a rotational mode, we found
the microdebrider’s oscillation mode to be more efficient in removing the lesion while preserving the
overlying epithelium. In addition, rather than setting
a fixed speed as recommended by Flint,6 we found
that using the variable speed controller allowed
more precise control over the resection. Our oscillation speeds of 800 to 1,000 revolutions per minute
were also higher than previously reported. We found
that the fibrous nature of the lesions did not allow
them to be easily resected at the lower speeds; however, we recommend using the lowest speed necessary to achieve resection.
One reported shortcoming of the microdebrider
is the inability to collect a specimen. Although the
amount of tissue removed is quite small, an in-line
specimen trap can be used to collect a specimen for
pathological examination. Certainly, if there are suspicious epithelial changes, a sample is obtained with
a small forceps and microscissors before the use of
the microdebrider. Another limitation of the microdebrider blade is its size. Although introduction
of the 2.9-mm blade was a major improvement, further refinement such as flattening of the tip would
allow easier insertion through the epithelial incision.
A case of anterior web and granuloma formation
after use of the microdebrider for removal of a polyp
has been reported.9 No such complications occurred
in our series. In our technique, the blade remains
subepithelial and the overlying surface is not violated. We do, however, agree that the highest level
of care is necessary in using powered instruments in
the larynx. In our opinion, they should not be used if
52
Cheng & Soliman, Subepithelial Microdebrider Excision
the lesion can be easily removed by traditional cold
microsurgical techniques.
this technique.
Although no formal voice analysis was performed,
all patients reported subjective improvement in their
voice after surgery; this was also evident on perceptual evaluation by the examining laryngologist.
Videostroboscopic examination of the larynx in the
early postoperative period showed findings typical
of those seen after the same procedure done without the microdebrider; they included edema, erythema, and stiffness of the operated vocal fold. These
findings were more pronounced in the patients with
Reinke’s edema, as expected, given that the incision
was larger and the dissection more extensive. Objective voice evaluation and comparison with traditional microsurgical techniques in a controlled study
is necessary to further determine the usefulness of
Microdebrider excision of subepithelial vocal fold
lesions is feasible. Patients with Reinke’s edema had
slightly more residual edema, a decreased mucosal
wave, and less improvement in voice as compared
to patients with pseudocysts. Meticulous technique,
low variable oscillatory speeds, and low wall suction
are necessary to avoid injury to the overlying epithelium and to the underlying superficial lamina propria and vocal ligament. Further refinements in instrument design would facilitate use through smaller
incisions. A larger, controlled study with objective
voice analysis and comparison with traditional microsurgical techniques is necessary to further define
the role of the microdebrider in the treatment of subepithelial vocal fold disease.
CONCLUSIONS
REFERENCES
1. Setliff RC III. The hummer: a remedy for apprehension
in functional endoscopic sinus surgery. Otolaryngol Clin North
Am 1996;29:95-104.
2. Myer CM III, Willging JP, McMurray S, Cotton RT. Use
of laryngeal micro resector system. Laryngoscope 1999;109:
1165-6.
3. Patel RS, Mackenzie K. Powered laryngeal shavers and
laryngeal papillomatosis: a preliminary report. Clin Otolaryngol
Allied Sci 2000;25:358-60.
4. Rees CJ, Tridico TI, Kirse DJ. Expanding applications for
the microdebrider in pediatric endoscopic airway surgery. Otolaryngol Head Neck Surg 2005;133:509-13.
5. Nuyens M, Zbaren P, Seifert E. Endoscopic resection of
laryngeal and tracheal lesions using the microdebrider. Acta
Otolaryngol 2006;126:402-7.
6. Flint P. Powered surgical instruments for laryngeal surgery. Otolaryngol Head Neck Surg 2000;122:263-6.
7. Sant’Anna GD, Mauri M. Use of the microdebrider for
Reinke’s edema surgery. Laryngoscope 2000;110:2214-6.
8. Honda K, Haji T, Maruyama H. Functional results of
Reinke’s edema surgery using a microdebrider. Ann Otol Rhinol Laryngol 2010;119:32-6.
9. Mortensen M, Woo P. An underreported complication of
laryngeal microdebrider: vocal fold web and granuloma: a case
report. Laryngoscope 2009;119:1848-50.
Response of Cricopharyngeus Muscle to Esophageal Stimulation
by Mechanical Distension and Acid and Bile Perfusion
Natalya Chernichenko, MD; Jeong-Soo Woo, MD; Jagdeep S. Hundal, MD;
Clarence T. Sasaki, MD
Objectives: The aim of this study was to identify the response of the cricopharyngeus muscle (CPM) to esophageal stimulation by intraluminal mechanical distension and intraluminal acid and bile perfusion.
Methods: In 3 adult pigs, electromyographic (EMG) activity of the CPM was recorded at baseline and after esophageal
stimulation at 3 levels: proximal, middle, and distal. The esophagus was stimulated with 20-mL balloon distension and
intraluminal perfusion of 40 mL 0.1N hydrochloric acid, taurocholic acid (pH 1.5), and chenodeoxycholic acid (pH 7.4)
at the rate of 40 mL/min. The EMG spike density was defined as peak-to-peak spikes greater than 10 μV averaged over
10-ms intervals.
Results: In all 3 animals, the spike density at baseline was 0. The spike densities increased after proximal and middle distensions to 15.2 ± 1.5 and 5.1 ± 1.2 spikes per 10 ms, respectively. No change in CPM EMG activity occurred after distal
distension. The spike density following intraluminal perfusion with hydrochloric acid at the distal level was 10.1 ± 1.1
spikes per 10 ms. No significant change in CPM EMG activity occurred after acid perfusion at the middle and proximal
levels. No change in CPM EMG activity occurred after intraluminal esophageal perfusion with either taurocholic acid or
chenodeoxycholic acid.
Conclusions: Proximal esophageal distension, as well as distal intraluminal acid perfusion, appeared to be important
mechanisms in generation of CPM activity. Bile acids, on the other hand, failed to evoke such CPM activity. The data
suggest that transpyloric refluxate may not be significant enough to evoke the CPM protective sphincteric function, thereby placing supraesophageal structures at risk of bile injury.
Key Words: bile acid, cricopharyngeus, gastroesophageal reflux.
INTRODUCTION
disease states is still incomplete. The CPM is tonically active at rest and relaxes during swallowing, belching, and vomiting. On the one hand, the
sphincteric function of the CPM remains controversial, perhaps serving to prevent esophageal distension during deep inspiration as intrathoracic pressure approaches subatmospheric, while volumetrically stabilizing the vocal tract by ensuring against
esophageal penetration as the vocal tract pressure
rises above atmospheric during phonation.3-5 On
the other hand, retrograde flow of esophageal contents has been increasingly recognized as a cause
of chronic lung disease and laryngeal dysfunction.6
Therefore, it has been suggested that evoked CPM
contraction in response to stimulatory events distal
to it may be protective against esophageal reflux and
aspiration.7 Others suggest that increased CPM tone
evoked by distal events provides the basis for hypo-
The upper esophageal sphincter (UES) is functionally defined as the high-pressure zone separating the pharynx from the cervical esophagus. Anatomically, the UES is a musculocartilaginous structure with its anterior wall defined by the posterior
surface of the cricoid cartilage inferiorly, as well as
the arytenoid and interarytenoid muscles superiorly.
Posteriorly and laterally, the UES is defined by the
inferior pharyngeal constrictor, the cricopharyngeus
muscle (CPM), and portions of the upper esophageal circular muscle fibers.1,2 It is generally believed, however, that the CPM is the main muscle
responsible for UES function.
Even though the anatomy of the UES is well defined, our current understanding of its normal physiologic function and its role in the development of
From the Section of Otolaryngology–Head and Neck Surgery, Yale University School of Medicine, New Haven, Connecticut. This
study was performed in accordance with the PHS Policy on Humane Care and Use of Laboratory Animals, the NIH Guide for the Care
and Use of Laboratory Animals, and the Animal Welfare Act (7 U.S.C. et seq.); the animal use protocol was approved by the Institutional Animal Care and Use Committee (IACUC) of Yale University.
Presented at the meeting of the American Broncho-Esophagological Association, Las Vegas, Nevada, April 28-29, 2010.
Correspondence: Clarence T. Sasaki, MD, Section of Otolaryngology–Head and Neck Surgery, Yale University School of Medicine,
333 Cedar St, PO Box 208041, New Haven, CT 06520.
53
54
Chernichenko et al, Cricopharyngeus Muscle in Esophageal Stimulation
pharyngeal pulsion diverticuli in susceptible populations by increasing hypopharyngeal pressure during swallowing, leading to herniation of the posterior pharyngeal wall weakness known as Killian’s
triangle. Thus, of all muscle groups in the head and
neck, none surpasses the CPM in capturing our clinical imagination and investigative curiosity.
Although the role of the UES has been studied
with the help of manometry, videofluoroscopy, videoendoscopy, and electromyographic (EMG) testing,8-10 the CPM’s contribution in terms of increased
muscle action potential in response to mechanical
distension, acid perfusion, and bile perfusion in the
same subject has not been demonstrated. This study
was designed to investigate the response of the CPM
to esophageal stimulation by mechanical distension,
hydrochloric acid perfusion, and bile acid perfusion.
Is its major sphincteric response due to intraluminal distension? Or does acid and/or bile perfusion
influence the EMG findings in the CPM? Does the
location of the stimulus within the esophagus alter
its response?
MATERIALS AND METHODS
Three adult male Yorkshire pigs were used in
the study. The animals were approximately 5 to 8
months of age, and weighed from 50 to 70 kg. The
porcine aerodigestive tract was selected as a model
because of its anatomic and physiologic similarity
to that of humans.11
Anesthesia was induced with an intramuscular injection of ketamine hydrochloride (30 mg/kg) and
xylazine hydrochloride (5 mg/kg) and was maintained with isoflurane inhalation via a nose cone.
By aseptic techniques, a vertical midline incision
was made extending from the level of the hyoid bone
to the sternal notch. A tracheotomy was performed
to provide maintenance inhalational anesthesia
through an endotracheal tube inserted through and
secured to the tracheostoma. This surgical exposure
also facilitated identification of the CPM and insertion of EMG electrodes. No pharmacologic muscle
relaxation was used. Electrocardiography, respiratory rate, pulse oximetry, and core body temperature
were continuously monitored and recorded every 15
minutes.
A 2-cm anterior pharyngotomy was performed
to allow direct visualization of the larynx and hypopharynx, as well as insertion of Foley and perfusion catheters under direct vision. Esophagoscopy
was performed via the pharyngotomy to establish
the total esophageal length. The esophageal length
was used to determine the level of proximal, middle,
and distal stimulation at 1 cm below the UES, 12
cm above the gastroesophageal junction, and 2 cm
above the gastroesophageal junction, respectively.
An XLTEX-Neuromax (Oakville, Canada) oscilloscope was used for EMG recording.
Mechanical Distension. A 12F Foley catheter was
used for mechanical distension of the esophagus. A
Foley catheter balloon was rapidly inflated with 20
mL of air delivered in 5 seconds to the distal, middle, and proximal parts of the esophagus in random
order. Each distension was separated in time by 5
minutes. The EMG activity was recorded at baseline
and with stimulation.
Intraluminal Perfusion With Hydrochloric Acid.
To simulate conditions of acid reflux, we perfused
the esophagus at the proximal, middle, and distal
levels with 40 mL of 0.1N hydrochloric acid (pH
1.5) at a rate of 40 mL/min via a Precision Syringe
Pump (Chemyx, Stafford, Texas) in random order.
Saline perfusion was used as a control. All perfusions were followed by a saline wash of 30 mL and
were separated by intervals of 5 minutes or until the
CPM EMG findings stabilized at baseline.
Intraluminal Perfusion With Bile Acids. To simulate conditions of bile reflux, we perfused the esophagus at the proximal, middle, and distal levels with
taurocholic acid (more than 95% pure; Sigma-Aldrich, St Louis, Missouri) at a concentration of 5
mmol/L titrated to pH 1.5 and chenodeoxycholic
acid (more than 98% pure; Sigma-Aldrich) at a concentration of 5 mmol/L titrated to a pH of 7.4 by
use of a protocol identical to that of the hydrochloric
acid perfusion.
A total of 6 EMG measures at each site were obtained for each protocol per animal to provide sufficient power for statistical analysis. The random
order of acid and bile application was intended to
reduce the possibility that intraluminal migration of
test solution might vertically contaminate the sitespecific level of esophageal stimulation. The EMG
spike density, which was defined as peak-to-peak
spikes greater than 10 μV averaged over 10-ms intervals, was calculated and compared to baseline.
The mean spike density and standard error were calculated.
RESULTS
In all 3 animals, the spike density at baseline was
0. Importantly, the 3 animals responded similarly to
stimulations. The CPM EMG activity occurred immediately after balloon distension. The spike densities following proximal and middle distensions increased and were 15.2 ± 1.5 and 5.1 ± 1.2 spikes
Chernichenko et al, Cricopharyngeus Muscle in Esophageal Stimulation
55
TABLE 1. SPIKE DENSITIES
Animal 1
Stimulation
Animal 2
Animal 3
Total
15.1 ± 3.9
0.5 ± 0.5
0
17.5 ± 6.1
11.8 ± 4.6
0
12.8 ± 5.6
3.0 ± 1.3
0
15.2 ± 1.5
5.1 ± 1.2
0
Following intraluminal perfusion with hydrochloric acid
Proximal
0
0
Middle
0
1.2 ± 0.8
Distal
0
12.8 ± 2.9
0
1.8 ± 0.7
8.2 ± 3.0
0
0.5 ± 0.2
9.2 ± 3.7
0
1.2 ± 0.4
10.1 ± 1.1
Baseline
Following esophageal distension
Proximal
0
Middle
0
Distal
0
Data are spikes per 10 ms (mean ± SE).
per 10 ms, respectively. No change in CPM EMG
activity occurred after distal distension as compared
to baseline (Table 1). Representative EMG recordings of CPM at baseline and following esophageal
distension at the proximal, middle, and distal levels
are presented in Fig 1.
The CPM EMG activity usually occurred 30 seconds after initiation of intraluminal perfusion of hydrochloric acid at the distal level (spike density, 10.1
± 1.1 spikes per 10 ms). No significant change in
Fig 1. Electromyographic (EMG) recordings of cricopharyngeus muscle (CPM) at baseline and following esophageal distension at proximal, middle, and distal levels.
Amplitude calibration is 10 μV. Time calibration is 10
ms.
CPM EMG activity occurred after perfusion at the
proximal and middle levels as compared to baseline (Table 1). Representative EMG recordings of
the CPM at baseline and following intraluminal perfusion with hydrochloric acid are presented in Fig
2. No change in CPM EMG activity occurred after
intraluminal esophageal perfusion with saline solution.
Notably, no change in CPM EMG activity occurred after intraluminal esophageal perfusion with
either taurocholic or chenodeoxycholic acid (Table
Fig 2. EMG recordings of CPM at baseline and following intraluminal perfusion with hydrochloric acid. Amplitude calibration is 10 μV. Time calibration is 10 ms.
56
Chernichenko et al, Cricopharyngeus Muscle in Esophageal Stimulation
TABLE 2. SPIKE DENSITY AT BASELINE AND
FOLLOWING INTRALUMINAL PERFUSION
WITH TC AND CDC
Proximal
Middle
Distal
TC
0
0
0
Baseline
CDC
0
0
0
Stimulation
TC
CDC
0
0
0
0
0
0
TC — taurocholic acid; CDC — chenodeoxycholic acid.
2). Representative EMG recordings of the CPM at
baseline and following intraluminal perfusion with
taurocholic and chenodeoxycholic acids are presented in Figs 3 and 4.
ry importance in mediating the UES relaxation response.9,13 Furthermore, the proximal rather than
the distal esophagus seems to be the stronger stimulus site for eliciting the esophageal-UES contractile
reflex in humans.14 This physiologic observation
finds morphological support in Neuhuber’s15 observation that in the rat model the most cranial part of
the esophagus receives the greatest vagal sensory
innervation and that the density of innervation decreases distally. Our functional EMG data from the
pig model support these observations.
Prior studies have clearly demonstrated that intraluminal esophageal balloon distension causes increased UES pressure.9,12 It has been also shown
that the esophageal-UES reflexes are mediated via
the brain stem by vagal afferents arising from the
esophageal mucosa and muscle layers. Mechanoreceptors located in the muscular layer are primarily
responsible for the contractile response of the UES,
whereas mucosal mechanoreceptors are of prima-
The role of intraluminal hydrochloric acid perfusion in the stimulation of CPM contraction has been
controversial. Gerhardt et al16 demonstrated a significant UES pressure increase when the proximal
esophagus of humans was stimulated with acid.
However, further studies have called this finding
into question by showing that the UES response to
intraluminal infusion of hydrochloric acid in normal
volunteers, although more pronounced at the proximal level, is indistinguishable from those produced
by saline solution and intraluminal distension, and
the investigators postulated that acidification may
not by itself stimulate this reflex.17 Still others have
Fig 3. EMG recordings of CPM at baseline and following
intraluminal perfusion with taurocholic acid. Amplitude
calibration is 10 μV. Time calibration is 10 ms.
Fig 4. EMG recordings of CPM at baseline and following
intraluminal perfusion with chenodeoxycholic acid. Amplitude calibration is 10 μV. Time calibration is 10 ms.
DISCUSSION
Chernichenko et al, Cricopharyngeus Muscle in Esophageal Stimulation
observed a significant rise in UES pressure following acid infusion 5 cm proximal to the gastroesophageal sphincter.18 We have clearly shown that in a
pig model hydrochloric acid stimulation of the distal esophagus induces CPM EMG activity, whereas
balloon distension at the same level elicits no change
in basal CPM activity. Therefore, it is reasonable to
conclude that acid stimulation in itself evokes CPM
activity, not merely enhancing the reflex response to
intraluminal distension.
It has previously been recognized that the earliest
change associated with injury from acid refluxate
is increased paracellular permeability due to damage to the intercellular junctions and subsequent development of “dilated intercellular spaces.”19 This
event allows hydrogen ions to penetrate the intercellular spaces and reach vagal sensory nerve endings
between the mucosal cells, in such a way as to trigger evoked CPM activity. One would expect hydrogen ions present in taurocholic acid solution at pH
1.5 to trigger a similar physiologic response. Interestingly, we observed no change in basal CPM activity in response to esophageal stimulation by bile
acids. Requiring further inquiry, the following hypothetical explanations are offered. On the one hand,
an absence of response to taurocholic acid (pKa 2)
could be attributed to its preferential “intramucosal
trapping” due to prevalence of its nonionized lipophilic form at pH 1.5 with subsequent entrapment
in the intracellular compartment due to ionization at
neutral pH.20 This “intramucosal trapping” of taurocholic acid may result in sufficient cellular edema to block paracellular permeability and therefore
prevent development of vagal stimulation related to
57
dilated intercellular spaces. On the other hand, it is
possible that the presence of taurocholic acid in addition to hydrogen ions could be so caustic as to either block or destroy sensory nerve endings, in such
a way as to block signal detection.
An absence of an evoked CPM response to esophageal stimulation by bile acids is an interesting observation in light of a growing understanding that
transpyloric refluxate in gastroesophageal reflux
is not uncommon. Although bile reflux may be directly injurious to esophageal and supraesophageal
mucosa,21 there does not seem to be sufficient evidence of its effect on evoked CPM sphincteric function. These observations further suggest that effective antacid therapy, while neutralizing the caustic
effects of acid on esophageal and supraesophageal
mucosa, may also blunt the effect of proximal CPM
protective function when gastroesophageal refluxate contains significant concentrations of caustic
bile salts at near-neutral pH, thereby placing supraesophageal structures at added risk of bile injury.
CONCLUSIONS
Physiologically, the cardinal events that occur
during gastroesophageal reflux are esophageal distension and reflux of acid with or without duodenal contents. Observed in our experiments, reflex
CPM contraction in response to proximal balloon
distension, as well as acid stimulation of the distal
esophagus, may be explained as a response acting
to prevent gastroesophageal refluxate from entering
the pharynx. However, transpyloric refluxate does
not appear a significant-enough stimulus to evoke a
CPM protective sphincteric function.
Acknowledgment: The authors are grateful to Dr Roy Orlando, MD, Department of Medicine, School of Medicine, University of
North Carolina, Chapel Hill, for his advice on acid-induced mucosal permeability.
REFERENCES
1. Sivarao DV, Goyal RK. Functional anatomy and physiology of the upper esophageal sphincter. Am J Med 2000;108(suppl 4A):27S-37S.
2. Singh S, Hamdy S. The upper oesophageal sphincter.
Neurogastroenterol Motil 2005;17(suppl 1):3-12. [Erratum in
Neurogastroenterol Motil 2005;17:773.]
3. Koufman JA. The otolaryngologic manifestations of gastroesophageal reflux disease (GERD): a clinical investigation
of 225 patients using ambulatory 24-hour pH monitoring and an
experimental investigation of the role of acid and pepsin in the
development of laryngeal injury. Laryngoscope 1991;101(suppl
53):1-78.
4. Sasaki CT, Kim YH, Sims HS, Czibulka A. Motor innervation of the human cricopharyngeus muscle. Ann Otol Rhinol
Laryngol 1999;108:1132-9.
5. Perera L, Kern M, Hofmann C, et al. Manometric evidence for a phonation-induced UES contractile reflex. Am J
Physiol Gastrointest Liver Physiol 2008;294:G885-G891.
6. Hunt PS, Connell AM, Smiley TB. The cricopharyngeus
sphincter in gastric reflux. Gut 1970;11:303-6.
7. Belsey R. Functional disease of the esophagus. J Thorac
Cardiovasc Surg 1966;52:164-88.
8. Shaker R, Ren J, Medda BK, et al. Identification and
characterization of the esophagoglottal closure reflex in a feline
model. Am J Physiol Gastrointest Liver Physiol 1994;266:G147G153.
9. Lang IM, Medda BK, Shaker R. Mechanism of reflexes
induced by esophageal distension. Am J Physiol Gastrointest
Liver Physiol 2001;281:G1246-G1263.
10. Lang IM, Dantas RO, Cook IJ, Dodds JW. Videoradiographic, manometric and electromyographic analysis of canine
upper esophageal sphincter. Am J Physiol Gastrointest Liver
Physiol 1991;260:G911-G919.
11. Schopf BW, Blair G, Dong S, Troger K. A porcine model
of gastroesophageal reflux. J Invest Surg 1997;10:105-14.
12. Creamer B, Schlegel J. Motor responses of the esophagus
to distension. J Appl Physiol 1957;10:498-504.
58
Chernichenko et al, Cricopharyngeus Muscle in Esophageal Stimulation
13. Szczesniak MM, Fuentealba SE, Burnett A, Cook IJ. Differential relaxation and contractile responses of the human upper esophageal sphincter mediated by interplay of mucosal and
deep mechanoreceptor activation. Am J Physiol Gastrointest
Liver Physiol 2008;294:G982-G988.
14. Enzmann DR, Harell GS, Zboralske FF. Upper esophageal responses to intraluminal distension in man. Gastroenterology 1977;72:1292-8.
15. Neuhuber WL. Sensory vagal innervation of the rat
esophagus and cardia: a light and electron microscopic anterograde tracing study. J Auton Nerv Syst 1987;20:243-55.
16. Gerhardt DC, Shuck TJ, Bordeaux RA, Winship DH. Human upper esophageal sphincter. Response to volume, osmotic,
and acid stimuli. Gastroenterology 1978;75:268-74.
17. Thompson DG, Andreollo NA, McIntyre AS, Earlam RJ.
Studies of the oesophageal clearance responses to intraluminal
acid. Gut 1988;29:881-5.
18. Wallin L, Boesby S, Madsen T. The effect of HCl infusion in the lower part of the esophagus on the pharyngo-esophageal sphincter pressure in normal subjects. Scand J Gastroenterol 1978;13:821-6.
19. Orlando RC. Current understanding of the mechanisms
of gastro-oesophageal reflux disease. Drugs 2006;66(suppl 1):
1-5, 29-33.
20. Schweitzer EJ, Bass BL, Batzri S, Harmon JW. Bile acid
accumulation by rabbit esophageal mucosa. Dig Dis Sci 1986;
31:1105-13.
21. Sasaki CT, Marotta J, Hundal J, Chow J, Eisen RN. Bileinduced laryngitis: is there a basis in evidence? Ann Otol Rhinol
Laryngol 2005;114:192-7.
Analysis of Pepsin in Tracheoesophageal Puncture Sites
Jonathan M. Bock, MD; Mary K. Brawley, MA; Nikki Johnston, PhD;
Tina Samuels, MS; Becky L. Massey, MD; Bruce H. Campbell, MD;
Robert J. Toohill, MD; Joel H. Blumin, MD
Objectives: Tracheoesophageal puncture (TEP) and prosthesis insertion is a well-established method of voice rehabilitation after laryngectomy. Maintenance of the prosthesis and tract can be challenging, and reflux to the TEP site has been
proposed as a cause. The sites of TEP were evaluated for the presence of pepsin in tissue biopsy specimens and tract secretions to explore this association.
Methods: Patients with TEP were interviewed for a history of symptoms related to reflux, medication use history, TEP
voice quality, and incidence of TEP complications. Tissue biopsy specimens and tract secretions were obtained from TEP
sites and analyzed for the presence of pepsin via sodium dodecyl sulfate–polyacrylamide gel electrophoresis Western
blot analysis.
Results: Twelve of 17 patients (47%) had some history of preoperative or postoperative symptoms of gastroesophageal
reflux disease or laryngopharyngeal reflux. Pepsin was present within the TEP site in a total of 10 of 17 patients (58%; 7
of 17 tissue biopsy specimens and 6 of 7 secretion samples). There were no statistically significant associations between
the presence of pepsin and sex, reflux history, use of acid suppressive medicine, or time since laryngectomy.
Conclusions: Reflux with subsequent pepsin deposition into the TEP tract occurs in a majority of laryngectomy patients.
Further studies on the effect of reflux on the health and function of the TEP tract are warranted.
Key Words: head and neck cancer, laryngectomy, pepsin, reflux, tracheoesophageal puncture.
method of TEP they described could be performed
in minutes as part of the initial laryngectomy or as
a secondary procedure. They also led efforts to develop easily replaceable prosthetic valves to facilitate adequate voice production and prevent backflow and aspiration of saliva. Over the past 30 years,
many improvements in valve prosthesis design have
been developed, but the problems of leakage, granulation, aspiration, debris, infection, and contamination continue to complicate prosthesis use. Required
rates of prosthesis change vary widely in reported
series, ranging from 2 months to 60 months.5
Over this same 30 years, information concerning
laryngopharyngeal reflux (LPR) and its relationship
to the development of laryngeal and pharyngeal cancers has appeared in the literature.6-11 Copper et al12
studied the prevalence of gastroesophageal reflux
disease (GERD) or LPR in 24 patients with laryngeal and pharyngeal squamous cell carcinomas. In
addition, 10 patients who had undergone irradiation
were evaluated for reflux. Using 24-hour dual-probe
pH monitoring, Copper et al12 determined that 20 of
24 cancer patients had pathological reflux, includ-
INTRODUCTION
Laryngectomy remains the standard of care for the
treatment of many advanced-stage or recurrent laryngeal cancers. Rehabilitation of alaryngeal speech
is a basic need for patients after this operation. The
idea of constructing a communication between the
upper trachea and pharynx for this purpose was developed in the 1960s, but initial attempts using this
technique for speech rehabilitation were complicated by problems of saliva and food leakage into
the trachea, as well as eventual stenosis of the tracheopharyngeal tract.1 Eventual efforts to surgically
create a communication tract between the pharynx
and trachea were pioneered by Asai,2 Miller,3 and
Montgomery and Toohill.1 These early procedures
were complicated by the need for multiple surgical
stages to create a tracheopharyngeal tract, and often
led to problems with tract stenosis, leakage, and aspiration.
The modern version of tracheoesophageal puncture (TEP) for postlaryngectomy speech was introduced by Singer and Blom4 in 1980. The endoscopic
From the Department of Otolaryngology and Communication Sciences, Medical College of Wisconsin, Milwaukee, Wisconsin.
Presented at the meeting of the American Broncho-Esophagological Association, Las Vegas, Nevada, April 28-29, 2010.
Correspondence: Jonathan M. Bock, MD, Dept of Otolaryngology and Communication Sciences, Medical College of Wisconsin, 9200
W Wisconsin Ave, Milwaukee, WI 53226.
59
60
Bock et al, Analysis of Pepsin in Tracheoesophageal Puncture Sites
TABLE 1. PATIENT REFLUX HISTORY QUESTIONNAIRE
Department of Otolaryngology and Communication Sciences
Center for Communication and Swallowing Disorders
Name: ___________________________________________
Date: ___________________________
Date of Laryngectomy: ______________________________
Date of TEP: _____________________
Before your laryngectomy, how did the following affect you?
(0 = no problem and 5 = severe problem)
1. Clearing your throat
0
1
2
2. Excess mucus or postnasal drip
0
1
2
3. Coughing after you eat or after lying down
0
1
2
4. Breathing difficulties or choking episodes
0
1
2
5. Troublesome or annoying cough
0
1
2
6. Sensations of something sticking in your throat or a lump in your throat
0
1
2
7. Heartburn, chest pain, indigestion, or stomach acid coming up
0
1
2
After your laryngectomy, how do the following affect you?
(0 = no problem and 5 = severe problem)
1. Excess mucus in your throat
0
1
2
2. Sensations of something sticking in your throat or a lump in your throat
0
1
2
3. Heartburn, chest pain, indigestion, or stomach acid coming up
0
1
2
Were you on reflux medication before your surgery?
_____yes
______no
Are you on reflux medication now?
_____yes
______no
Medication: ______________________________________
Subject number: _______
3
3
3
3
3
3
3
4
4
4
4
4
4
4
5
5
5
5
5
5
5
3
3
3
4
4
4
5
5
5
ing 62% with LPR on testing. They concluded that
LPR is a common problem in patients with head and
neck cancer.12
the TEP site.
Ongoing reflux into the upper esophagus after laryngectomy would not be particularly bothersome if
there were no communication between the esophagus and the airway. However, when TEP has been
performed for voice rehabilitation, this communication may be more vulnerable to the effects of reflux,
as some of the protective mechanisms that retard reflux may be disturbed by laryngectomy. A study by
Pattani et al13 confirmed that acid suppression therapy was beneficial to the majority of TEP patients as
it related to voice production and TEP health. Studies have also indicated that pepsin can be injurious
to the extraesophageal tissues, suggesting that acid
reduction therapy using proton pump inhibitors may
not be sufficient to inhibit damage to these areas, including the TEP site.14
Patients. This study was approved by the Institutional Review Board of the Medical College of Wisconsin. The study volunteers were initially identified from the TEP registry of the speech-language
pathology section of the Department of Otolaryngology and Communication Sciences of the Medical
College of Wisconsin. All patients used TEP prostheses from Atos Medical Inc (Hörby, Sweden). All
consecutive laryngectomy patients were included in
the study as they presented for routine TEP maintenance. Enrolled patients were interviewed for past
evidence of LPR and/or GERD before and after laryngectomy (Table 1). Use of acid suppressive medication before and after surgery was also documented. Symptoms of LPR and GERD were judged on a
scale from 0 (none) to 5 (severe) on the basis of average scores on survey analysis. Frequency of prosthesis changes and the presence of granulation, leakage, or prosthetic debris were noted.
Detection of pepsin in tissues or fluid secretions
is a relatively simple and noninvasive method to detect ongoing reflux events to extraesophageal structures.14 We hypothesize that many observed difficulties with TEP, such as frequent prosthesis changes
or poor alaryngeal speech, may be associated with
reflux of gastric contents to the TEP site. The purpose of this study was to evaluate the prevalence of
reflux in patients who had undergone laryngectomy
and TEP placement by analysis for pepsin in biopsy
specimens from the TEP tract and secretions from
METHODS
Specimens. Biopsy specimens of the TEP tract
were obtained along the tract with a 2-mm cup forceps (Fig 1) and placed in sterile saline solution until later analysis. Secretions were obtained by collecting fluid from around the TEP tract site into a
small suction trap (No. 7419101, Medtronic Xomed,
Jacksonville, Florida).
Voice Analysis. Enrolled study participants had
Bock et al, Analysis of Pepsin in Tracheoesophageal Puncture Sites
61
and secretion results and clinical parameters (GERD
and LPR symptoms, current proton pump inhibitor
therapy, gender, time since laryngectomy, history of
radiotherapy) were compared by use of Fisher’s exact test. Associations between pepsin status and TEP
change rate and complications were analyzed by use
of Student’s t-test. Statistical significance was established at a p value of less than 0.05.
RESULTS
Fig 1. Tissue biopsy specimens were obtained from
tracheoesophageal puncture (TEP) tract sites by use of
2-mm cup forceps at middle portion of tract. Associated
secretions were collected in suction trap.
their TEP voicing interpreted by an unblinded
speech-language pathologist (M.K.B.), and overall
voice quality was assessed by interpreting vocal fluency, mucoid wetness, strain, and breathiness to formulate an overall impression of TEP voice function
(good, fair, poor).
Western Blotting. Total protein was extracted
from biopsy specimens and secretions from the TEP
tract in urea lysis buffer,15 and protein content was
measured by Bradford assay (No. 500-0006, BioRad, Hercules, California). We loaded 30 μg of total
protein (less in the case of low-concentration specimens) on 10% sodium dodecyl sulfate–polyacrylamide gel electrophoresis gels according to standard
protocol. Purified human pepsin 3b16 and human
pepsinogen I (P1490, Sigma, St Louis, Missouri)
were run alongside TEP samples as positive and
negative controls, respectively. Proteins were transferred to polyvinylidene fluoride membrane (RPN2020F, GE Healthcare, Piscataway, New Jersey)
and probed with rabbit antipepsin antibody (1:35014)
and mouse anti-β-actin antibody (1:5000, No. CP01,
EMD Chemicals, Gibbstown, New Jersey). Blots
were then probed with appropriate peroxidase-conjugated secondary antibody diluted 1:5,000 (P0447/
P0448, Dako, Copenhagen, Denmark). All antibodies were diluted in phosphate-buffered saline solution, 0.1% Tween-20, and 5% nonfat dry milk. Blots
were exposed to enhanced chemiluminescence reagents (sc-2048, Santa Cruz Biotechnology, Santa
Cruz, California) and then submitted to radiographic
exposure and development.
Statistics. Associations between pepsin biopsy
Seventeen patients who had undergone laryngectomy with TEP (ages 51 to 76 years; mean, 66 years;
12 male, 5 female) were enrolled in this study between September 2009 and January 2010 (Table 2).
The average length of time since laryngectomy was
39 months (range, 6 to 129 months), and the length
of time since TEP placement was 37.6 months
(range, 6 to 121 months). The average time between
prosthesis changes was 1.97 months (range, 0.5 to
4.8 months). Most patients (11 of 17, or 65%) had
a TEP placed as a primary procedure, and the remainder had their TEP placed secondarily within 3
to 6 months after laryngectomy. Most patients (13
of 17, or 76%) had a history of irradiation during
their cancer treatment. The most common reason for
prosthesis change was leakage around the prosthesis. Voicing was judged as good in 13 patients, fair
in 2 patients, and poor in 2 patients.
All patients had biopsy specimens taken of tissue from the TEP tract. Seven of these biopsy specimens (41%) showed positive sodium dodecyl sulfate–polyacrylamide gel electrophoresis immunoblotting for the presence of pepsin (Fig 2A). Later
in the study, we also collected a concurrent specimen of the associated secretions from the TEP tract
as described above. Three patients had these secretions tested for pH, and all were found to have secretions of pH 6.5 to 7. Six of 7 TEP secretion specimens (85%) had positive immunoblot staining for
the presence of pepsin (Fig 2B). One half of patients
with secretions demonstrating pepsin also had biopsy specimens that were positive for pepsin. A total of
10 of 17 patients (59%) tested positive for pepsin in
either the TEP tract biopsy specimen or secretions.
Nine patients had prelaryngectomy symptoms
suggestive of LPR (excessive mucus, globus sensation, acid taste), and these symptoms persisted in
all 9 after laryngectomy, although there was a general trend toward decreased severity of LPR symptoms. Symptoms of GERD (including heartburn,
chest pain, and indigestion) were present in 6 cases
before laryngectomy, and all of these patients continued to note these problems after laryngectomy.
Two patients who denied preoperative symptoms of
62
Bock et al, Analysis of Pepsin in Tracheoesophageal Puncture Sites
TABLE 2. PATIENT DATA
TL
Pt Age
Duration
No. (y) Sex
(mo)
RT
1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
52
F
87
–
LPR
Before
TL
Severe
LPR
After
TL
Mild
GERD
Before
TL
Average
GERD PPI
PPI
TEP
Pepsin Pepsin
After Before After Change
Voice
in TEP in TEP
TL
TL
TL Rate (mo) Quality Complications Biopsy Secretions
Severe Severe
Debris,
–
granulation,
leakage
71 F
82
+ Moderate Moderate
–
–
+
+
4.0
Poor
Debris,
+
leakage
51 M
12
+ Mild
Mild
–
–
–
+
0.5
Good
Debris,
–
+
leakage
77 M
6
+ Mild
Moderate Mild
Mild
+
+
1.0
Fair
Leakage
–
74 M
10
+ Mild
Moderate
–
–
–
+
0.6
Good
Leakage
+
51 F
11
+ Severe
Mild
Severe Severe
+
+
1.2
Good
Leakage
+
74 F
39
+
–
–
Severe Severe
+
+
1.8
Good
Leakage
–
73 M
31
+ Severe
Mild
Severe Severe
+
+
1.6
Good
Debris,
–
leakage
75 M
42
+
–
–
–
–
–
+
4.8
Poor
Leakage
–
66 M
85
+
–
–
–
Severe
–
+
1.7
Good
Debris,
+
+
leakage
76 M
129
–
–
–
–
–
–
+
1.0
Good
None
+
59 M
107
+
–
–
–
–
–
+
2.4
Good
Debris,
+
+
leakage
59 M
33
+
–
–
–
–
–
–
1.5
Good
Debris,
+
+
leakage
75 M
9
+
–
–
–
–
–
–
2.5
Fair
Leakage
–
+
65 M
16
+
–
–
–
Moderate –
–
1.8
Good
Debris,
–
–
leakage
67 M
42
– Mild
Mild
–
–
–
–
4.2
Good
Leakage
–
52 F
121
– Severe
Severe
Severe Severe
+
+
1.7
Good
Debris,
–
+
granulation,
leakage
TL — total laryngectomy; RT — radiotherapy; LPR — laryngopharyngeal reflux; GERD — gastroesophageal reflux disease; PPI — proton pump
inhibitor; TEP — tracheoesophageal puncture.
GERD noted them after laryngectomy, leading to a
total of 8 patients (47%) with postoperative symptoms of GERD. There was no statistical association
between pepsin positivity of the tract biopsy specimen and current GERD symptoms (p = 0.18, Fisher
exact test). Acid suppressive medications were commonly used in our study group, but were unrelated to
+
+
1.3
Good
pepsin positivity of the tract biopsy (p = 0.4, Fisher
exact test). No statistically significant associations
between the presence of pepsin and the frequency of
prosthesis changes, history of radiotherapy, or incidence of various TEP complications could be made
on the basis of this limited observational patient series (p = 0.6, Student’s t-test).
Fig 2. Western blotting data. Biopsy specimens and associated secretions from TEP tract were assayed for presence of pepsin
by standard sodium dodecyl sulfate–polyacrylamide gel electrophoresis techniques. Proteins were separated by electrophoresis,
transferred to nitrocellulose membranes, and probed for pepsin and β-actin. Purified human pepsinogen 1 and human pepsin
were included as negative and positive immunoblotting controls, respectively. Representative TEP biopsy extracts are shown
(TEP11 to TEP14) with range of signal strength, including one negative assay (TEP14) with strong β-actin signal. Representative TEP secretion assays are shown on right (TEP12S, TEP14S, and TEP15S), again showing range of intensity with negative
sample (TEP15S).
Bock et al, Analysis of Pepsin in Tracheoesophageal Puncture Sites
DISCUSSION
Both acidic and nonacidic elements of gastric
refluxate, including pepsin, remain a therapeutic
problem in many patients with reflux, and the TEP
population is no exception. Current work with impedence technology is attempting to establish better normative data for reflux events in normal and
pathological conditions,17 but the overall effect of
digestive enzyme deposition on laryngeal tissues
is still an area of active research. Pepsin is a proteolytic enzyme produced only by the mucosa of
the stomach, and detection of this protein outside
the stomach provides evidence of ongoing reflux of
stomach contents.18 We have previously identified
that esophageal mucosa does not significantly uptake pepsin, even in patients with pH-probe–proven
LPR, whereas laryngeal mucosa seems much more
sensitive to pepsin uptake.19 Identification of pepsin
in the TEP tract and associated secretions therefore
suggests the presence of reflux episodes into the upper esophagus in our study subjects, and our data
support the use of pepsin analysis of TEP biopsy
specimens and secretions for detection of ongoing
reflux to the TEP site. It is unclear whether secretion testing alone is sufficient to predict the possible
detrimental tissue effects from pepsin at this time;
however, the noninvasive nature of secretion testing
is favorable for clinical use. Studies to evaluate the
mechanism of pepsin uptake into tissues of the larynx and esophagus are necessary to clarify the effect
of pepsin in secretions on the adjacent tissues. Pepsin uptake appears to be tissue-specific, and is seen
in the esophagus and larynx of patients with LPR,
but is not present in nasopharyngeal or palatine tonsil biopsy specimens. Patients with LPR also often
have pepsin present in nasopharyngeal secretions
despite negative nasopharyngeal tissue biopsies (N.
Johnston, unpublished data). Pepsin assays hold significant promise for detecting reflux, and noninvasive testing for pepsin may become more widely
accepted in the future.18 There are no commercial
methods for testing for the presence of pepsin in
oropharyngeal or esophageal secretions at this time,
but such products are in development and promise
to greatly enhance the care of patients with refluxassociated disorders.
The management of nonacidic reflux is incompletely addressed with currently available treatments.
Antireflux surgery, including Nissen fundoplication,
is likely the best option at present for total control of
esophageal reflux. Studies have shown good control
of extraesophageal symptoms from reflux in carefully selected patients known to have LPR20; however, the invasive nature of this intervention always
requires careful consideration. Few studies have in-
63
vestigated the role of fundoplication in the control
of reflux after laryngectomy. Jobe et al21 published a
series of 9 patients with laryngectomy and TEP who
were treated with fundoplication for known reflux,
and described severe problems with bloating and
hyperflatulence after fundoplication in this cohort.
Given these surgical challenges, improved medical treatment protocols would be of great benefit to
these patients. Liquid alginate suspension is an effective antireflux medication that is currently available in Europe (Gaviscon Advance, Reckitt Benckiser Healthcare, Kingston-Upon-Thames, England)
and holds great promise as a potential treatment for
both acidic and nonacidic reflux.22 It is a well-tolerated mucosal protectant with minimal side effects as
compared to acid suppressive therapy.23 Other options for treatment could include gastric prokinetic agents such as metaclopramide or erythromycin,
although long-term use of these agents in patients
with reflux remains controversial.24
Just over half of the patients in our study had
symptomatic complaints of LPR before the diagnosis of laryngeal cancer, and all of these patients
continued to have pharyngeal symptoms of reflux
after laryngectomy, including globus sensation, excessive mucus, and throat irritation. A third of patients had persistent symptoms of GERD after surgery. There was a mixed association between GERD
and LPR symptoms and the presence of pepsin in
the TEP tract. The majority of the TEP patients studied in this protocol were treated with acid suppression medications, including 8 of the 10 patients who
had positive results for pepsin. The usual problems
of granulation, leakage, prosthetic debris, contamination, and the need for frequent prosthesis changes
were also apparent in this patient population. The
majority of the patients included in this study had a
frequent need for prosthetic change (average, 1.97
months), and perhaps this study was biased to those
with more severe or ongoing issues related to TEP
speech and a heightened risk of LPR. Nearly all patients in this study had some degree of complications from TEP placement. However, only 2 of the
17 patients in our cohort had TEP voicing problems
that were significant enough to be noted by others.
One of these patients was noted to have pepsin in
the TEP tract with multiple associated TEP complications, but the other did not have pepsin present
and had minimal complications. These data provide
some further support for the recommendation that
TEP patients should be routinely treated with acid
suppression medications.13
The effects of laryngectomy and reflux on the
overall function of the digestive tract remain unclear
at this time, and many potential questions arise in
64
Bock et al, Analysis of Pepsin in Tracheoesophageal Puncture Sites
considering this issue. Laryngectomy is known to
cause increased esophageal transit time, decreased
esophageal contraction strength, altered proximal
esophageal contraction, and deranged upper esophageal sphincter opening.25 The effects of laryngectomy on the function of the lower esophageal sphincter are less clear. The direct effect of regurgitation
of gastric juices into the larynx is a well-known entity, but is there an added reflex, likely via the vagus nerve or autonomic nervous system, that promotes reflux through the lower esophageal sphincter.26 Does laryngectomy alter lower esophageal
sphincter function via derangement of this vagal
pathway once the superior and recurrent laryngeal
nerves are transected? Few studies have directly addressed these questions, but some data exist to show
that function of the lower esophageal sphincter remains intact after laryngectomy or laryngeal irradiation.27,28 The ultimate alteration of the larynge-
al function and physiology occurs with total laryngectomy, and evidence indicates that laryngectomy
may prolong or extend the impact of preoperative
reflux symptoms.21,25,29,30 The role that laryngectomy plays in altering esophageal physiology and subsequent reflux warrants continued study.
In conclusion, we demonstrated in this observational study that pepsin is present in the TEP tract
tissues or secretions of more than half (10 of 17,
or 58%) of laryngectomy patients, suggesting that
a majority of these patients have ongoing reflux.
Acid suppression appears to have little impact on
the presence of pepsin in the secretions and tissues
of the TEP tract of laryngectomy patients. Our data
suggest that further studies are warranted of the impact of pepsin and reflux on the mucosal health and
function of the extraesophageal tissues in patients
who have undergone TEP and laryngectomy.
REFERENCES
1. Montgomery WW, Toohill RJ. Voice rehabilitation after
laryngectomy. Arch Otolaryngol 1968;88:499-506.
2. Asai R. Asai’s new voice production method: a substitution for human speech. In: Proceedings of the 8th International
Congress of Otorhinolaryngology, Tokyo, Japan, 1965.
3. Miller AH. First experiences with the Asai technique for
vocal rehabilitation after total laryngectomy. Ann Otol Rhinol
Laryngol 1967;76:829-33.
4. Singer MI, Blom ED. An endoscopic technique for restoration of voice after laryngectomy. Ann Otol Rhinol Laryngol
1980;89:529-33.
5. Ferrer Ramírez MJ, Guallart Doménech F, Brotons Durbán S, Carrasco Llatas M, Estellés Ferriol E, López Martínez R.
Surgical voice restoration after total laryngectomy: long-term
results. Eur Arch Otorhinolaryngol 2001;258:463-6.
6. Glanz H, Kleinsasser O. Chronic laryngitis and carcinoma [in German]. Arch Otorhinolaryngol 1976;212:57-75.
7. Ward PH, Hanson DG. Reflux as an etiological factor of
carcinoma of the laryngopharynx. Laryngoscope 1988;98:11959.
8. Morrison MD. Is chronic gastroesophageal reflux a causative factor in glottic carcinoma? Otolaryngol Head Neck Surg
1988;99:370-3.
9. Koufman JA. The otolaryngologic manifestations of gastroesophageal reflux disease (GERD): a clinical investigation
of 225 patients using ambulatory 24-hour pH monitoring and an
experimental investigation of the role of acid and pepsin in the
development of laryngeal injury. Laryngoscope 1991;101(suppl
53):1-78.
10. Freije JE, Beatty TW, Campbell BH, Woodson BT,
Schultz CJ, Toohill RJ. Carcinoma of the larynx in patients with
gastroesophageal reflux. Am J Otolaryngol 1996;17:386-90.
11. Biacabe B, Gleich LL, Laccourreye O, Hartl DM, Bouchoucha M, Brasnu D. Silent gastroesophageal reflux disease in
patients with pharyngolaryngeal cancer: further results. Head
Neck 1998;20:510-4.
12. Copper MP, Smit CF, Stanojcic LD, Devriese PP, Schou-
wenburg PF, Mathus-Vliegen LM. High incidence of laryngopharyngeal reflux in patients with head and neck cancer. Laryngoscope 2000;110:1007-11.
13. Pattani KM, Morgan M, Nathan CO. Reflux as a cause
of tracheoesophageal puncture failure. Laryngoscope 2009;119:
121-5.
14. Johnston N, Knight J, Dettmar PW, Lively MO, Koufman J. Pepsin and carbonic anhydrase isoenzyme III as diagnostic markers for laryngopharyngeal reflux disease. Laryngoscope
2004;114:2129-34.
15. Axford SE, Sharp N, Ross PE, et al. Cell biology of laryngeal epithelial defenses in health and disease: preliminary
studies. Ann Otol Rhinol Laryngol 2001;110:1099-108.
16. Crapko M, Kerschner JE, Syring M, Johnston N. Role of
extra-esophageal reflux in chronic otitis media with effusion.
Laryngoscope 2007;117:1419-23.
17. Savarino E, Tutuian R, Zentilin P, et al. Characteristics
of reflux episodes and symptom association in patients with
erosive esophagitis and nonerosive reflux disease: study using
combined impedance–pH off therapy. Am J Gastroenterol 2010;
105:1053-61.
18. Samuels TL, Johnston N. Pepsin as a marker of extraesophageal reflux. Ann Otol Rhinol Laryngol 2010;119:203-8.
19. Johnston N, Dettmar PW, Lively MO, et al. Effect of pepsin on laryngeal stress protein (Sep70, Sep53, and Hsp70) response: role in laryngopharyngeal reflux disease. Ann Otol Rhinol Laryngol 2006;115:47-58.
20. Catania RA, Kavic SM, Roth JS, et al. Laparoscopic Nissen fundoplication effectively relieves symptoms in patients with
laryngopharyngeal reflux. J Gastrointest Surg 2007;11:157988.
21. Jobe BA, Rosenthal E, Weisberg TT, et al. Surgical management of gastroesophageal reflux and outcome after laryngectomy in patients using tracheoesophageal speech. Am J Surg
2002;183:539-43.
22. McGlashan JA, Johnstone LM, Sykes J, Strugala V, Dettmar PW. The value of a liquid alginate suspension (Gaviscon
Bock et al, Analysis of Pepsin in Tracheoesophageal Puncture Sites
Advance) in the management of laryngopharyngeal reflux. Eur
Arch Otorhinolaryngol 2009;266:243-51.
23. Hampson FC, Jolliffe IG, Bakhtyari A, et al. Alginateantacid combinations: raft formation and gastric retention studies. Drug Dev Ind Pharm 2010;36:614-23.
24. Emerenziani S, Sifrim D. Gastroesophageal reflux and
gastric emptying, revisited. Curr Gastroenterol Rep 2005;7:1905.
25. Choi EC, Hong WP, Kim CB, et al. Changes of esophageal motility after total laryngectomy. Otolaryngol Head Neck
Surg 2003;128:691-9.
26. Mittal RK, Balaban DH. The esophagogastric junction.
N Engl J Med 1997;336:924-32.
65
27. Dantas RO, Aguiar-Ricz LN, Oliveira EC, Mello-Filho
FV, Mamede RC. Influence of esophageal motility on esophageal
speech of laryngectomized patients. Dysphagia 2002;17:121-5.
28. Gaze MN, Wilson JA, Gilmour HM, MacDougall RH,
Maran AG. The effect of laryngeal irradiation on pharyngoesophageal motility. Int J Radiat Oncol Biol Phys 1991;21:131520.
29. Welch RW, Luckmann K, Ricks PM, Drake ST, Gates
GA. Manometry of the normal upper esophageal sphincter and
its alterations in laryngectomy. J Clin Invest 1979;63:1036-41.
30. Gerwin JM, Culton GL, Gerwin KS. Hiatal hernia and reflux complicating prosthetic speech. Am J Otolaryngol 1997;18:
66-8.
Low-Acid Diet for Recalcitrant Laryngopharyngeal Reflux:
Therapeutic Benefits and Their Implications
Jamie A. Koufman, MD
Objectives: Laryngopharyngeal reflux (LPR) is an expensive, high-prevalence disease with a high rate of medical treatment failure. In the past, it was mistakenly believed that pepsin was inactive above pH 4; however, human pepsin has
been reported to be active up to pH 6.5. In addition, it has been shown by Western blot analysis that laryngeal biopsy samples from patients with symptomatic LPR have tissue-bound pepsin. The clinical impact of a low-acid diet on the therapeutic outcome in LPR has not been previously reported. To provide data on the therapeutic benefit of a strict, virtually
acid-free diet on patients with recalcitrant, proton pump inhibitor (PPI)–resistant LPR, I performed a prospective study of
20 patients who had persistent LPR symptoms despite use of twice-daily PPIs and an H2-receptor antagonist at bedtime.
Methods: The reflux symptom index (RSI) score and the reflux finding score (RFS) were determined before and after
implementation of the low-acid diet, in which all foods and beverages at less than pH 5 were eliminated for a minimum
2-week period. The subjects were individually counseled, and a printed list of acceptable foods and beverages was provided.
Results: There were 12 male and 8 female study subjects with a mean age of 54.3 years (range, 24 to 72 years). The
symptoms in 19 of the 20 subjects (95%) improved, and 3 subjects became completely asymptomatic. The mean pre-diet
RSI score was 14.9, and the mean post-diet RSI score was 8.6 (p = 0.020). The mean pre-diet RFS was 12.0, and the mean
post-diet RFS was 8.3 (p < 0.001).
Conclusions: A strict low-acid diet appears to have beneficial effects on the symptoms and findings of recalcitrant (PPIresistant) LPR. Further study is needed to assess the optimal duration of dietary acid restriction and to assess the potential
role of a low-acid diet as a primary treatment for LPR. This study has implications for understanding the pathogenesis,
cell biology, and epidemiology of reflux disease.
Key Words: acid reflux, adenocarcinoma, antireflux, Barrett’s esophagus, chronic cough, diet, esophageal cancer, gastroesophageal reflux disease, heartburn, hoarseness, laryngopharyngeal reflux, low acid, low fat, pepsin, proton pump
inhibitor.
jury — and it is peptic (not acid) injury.1,5,8,9,11-17
(Pepsin does, however, require some acid for activation.) We previously showed that 19 of 20 patients
(95%) with clinical and pH-documented LPR had
tissue-bound pepsin identifiable by Western blot
analysis, as opposed to only 1 of 20 control subjects
(5%).9 In addition, peptic injury is associated with
depletion of key protective proteins, including carbonic anhydrase, E-cadherin, and most of the stress
proteins.5,8,9,11-15
INTRODUCTION
Laryngopharyngeal reflux (LPR) is a controversial, high-prevalence disease, and it differs from
classic gastroesophageal reflux disease (GERD) in
many ways.1-10 Typically, patients with LPR have
daytime (upright) reflux without having heartburn
or esophagitis.1-3 In addition, one of the most important differences between LPR and GERD is that
the threshold for laryngeal tissue damage is much
lower than that for the esophagus.1,5,8 As many as
50 reflux episodes (less than pH 4) per day are considered normal for the esophagus, whereas as few
as 3 reflux episodes per week are too many for the
larynx.1
Equally important in understanding the biology
of LPR is consideration for the stability and spectrum of activity of human pepsin.14 In the past, it
was mistakenly believed that pepsin was inactive
above pH 4.1 The early experiments on which that
result was based were performed with porcine pepsin, and not human pepsin. Indeed, pig pepsin is inactive at greater than pH 4; however, human pepsin
retains some of its proteolytic activity up to pH 6.5,
THERAPEUTIC IMPLICATIONS OF CELL BIOLOGY
OF LPR
The cell biology of LPR holds the key to understanding the susceptibility of the larynx to peptic in-
From the Voice Institute of New York, New York, NY.
Presented at the meeting of the American Broncho-Esophagological Association, Las Vegas, Nevada, April 28-29, 2010.
Correspondence: Jamie A. Koufman, MD, Voice Institute of New York, 200 W 57th St, Suite 1203, New York, NY 10019.
66
Koufman, Low-Acid Diet for Laryngopharyngeal Reflux
67
TABLE 1. TRADITIONAL ANTIREFLUX DIET AND
LIFESTYLE MODIFICATION PROGRAM
Human pepsin activity curve. (Data from Johnston et
al.14)
depending on the substrate.14 The pepsin activity
curve is shown in the Figure.14 Peak peptic activity
(100%) occurs at pH 2, but there is still some (10%)
activity at pH 6.14 In other words, clinical disease
(LPR) is associated with tissue-bound pepsin,9 and
laryngeal damage occurs at pH 5.0 or less.8
CLINICAL CONSIDERATIONS
For more than 25 years, LPR was diagnosed in
my practice by the symptoms and findings of LPR
and by ambulatory 24-hour (simultaneous pharyngeal and esophageal) pH monitoring.1-3,7 Treatment
for moderate to severe LPR was typically twice-daily proton pump inhibitors (PPIs) with an H2-receptor antagonist at bedtime and an antireflux dietary
and lifestyle modification program (Table 1).
There was some customization of the conventional antireflux protocol, eg, one cup of coffee a day,
no citrus, no carbonated beverages. We have long
recognized that some patients who drank excessive
amounts of carbonated beverages might gain control
of their LPR simply by eliminating those beverages. Indeed, carbonated beverage consumption is one
of the most common identifiable causes of medical
treatment failure in LPR.
With our 2007 study14 showing peptic activity
up to pH 6.5, and having previously found (by immunohistochemical analysis) pepsin within the tissue biopsy specimens of patients with LPR,9 we
recognized that tissue-bound pepsin in these patients might be activated by exogenous hydrogen
ions from any source, including dietary ones. Consequently, in 2008 we began measuring the pH of
common foods and beverages and restricting patients with LPR from consuming anything below
pH 5 for a trial period of 2 weeks. To our surprise,
this appeared to have outstanding therapeutic benefits for many patients.
In the ensuing few years, we continued to refine
If you use tobacco, quit, because smoking causes reflux.
Do not wear clothing that is too tight, especially trousers,
corsets, and belts.
Avoid exercising, especially weight-lifting, swimming,
jogging, and yoga, after eating.
Do not lie down after eating; do not eat within 3 hours of
bedtime.
Elevate the head of your bed if you have nighttime reflux
(hoarseness, sore throat, and/or cough in the morning).
Limit your intake of red meat, butter, cheese, eggs, and
anything with caffeine.
Completely avoid fried food, high-fat meats, onions, tomatoes,
citrus fruit or juice, carbonated beverages (soda), beer, hard
liquor, wine, mints, and chocolate.
this induction reflux diet to exclude all recognized
reflux trigger foods, as well as anything that we identified to be acidic (below pH 5). Eggs and red apples,
for example, used to be on the induction diet, but
we found that those foods caused problems for some
of our patients, so they were removed. Thus, the approved foods and beverages list for the induction reflux diet evolved to its present form (Table 2).
MATERIALS AND METHODS
All of my adult patients with pH-documented
LPR on “maximum” antireflux therapy (twice-daily
PPIs with an H2-receptor antagonist at night) were
eligible for the study if they were failing to improve
on medical treatment and did not have potentially
life-threatening manifestations of LPR. Specifically
excluded were patients with airway stenosis, laryngeal neoplasia, and/or pulmonary disease. Medical
treatment failure was defined as no improvement
in symptoms — according to a validated outcomes
measure, the reflux symptom index (RSI)18,19 —
on office visits at least 2 months apart. Incidentally, it has been my routine practice for the past 25
years to have all patients complete the RSI at every
visit.
Before inclusion in the low-acid diet study, patients were offered other alternatives such as changing their antireflux medications or evaluation for
antireflux surgery. If the low-acid reflux diet was
elected (Table 2), patients had to agree to be compliant with the conditions of the diet, which was very
restrictive (nothing below pH 5). For example, the
diet allows no fruit except bananas and melons. Patients were counseled about what they could and
could not eat and were provided with a handout that
explained the purpose of the diet and its scientific
basis, as well as a list of foods and beverages that
were allowed. Study subjects were instructed to eat
only from the list of the items in Table 2 for 2 weeks
68
Koufman, Low-Acid Diet for Laryngopharyngeal Reflux
TABLE 2. INDUCTION REFLUX DIET
Agave
Aloe vera
Artificial sweetener (maximum 2 teaspoons per day)
Bagels and (non-fruit) low-fat muffins
Banana (great snack food)
Beans (black, red, lima, lentils, etc)
Bread (especially whole grain and rye)
Caramel (maximum 4 tablespoons per week)
Celery (great snack food)
Chamomile tea (most other herbal teas are not acceptable)
Chicken (grilled, broiled, baked, or steamed; no skin)
Chicken stock or bouillon
Coffee (maximum 1 cup per day; best with milk)
Egg whites
Fennel
Fish (grilled, broiled, baked, or steamed)
Ginger (ginger root, powdered, or preserved)
Graham crackers
Herbs (excluding all peppers, citrus, garlic, and mustard)
Honey
Melon (honeydew, cantaloupe, watermelon)
Mushrooms (raw or cooked)
Oatmeal (all whole-grain cereals)
Olive oil (maximum 2 tablespoons per day)
Parsley
Pasta (with nonacidic sauce)
Popcorn (plain or salted, no butter)
Potatoes (all of the root vegetables except onions)
Rice (healthy, especially brown rice, a staple during induction)
Skim milk (alternatively, soy or Lactaid skim milk)
Soups (great homemade with noodles and vegetables)
Tofu
Turkey breast (organic, no skin)
Turnip
Vegetables (raw or cooked, but no onion, tomato, or peppers)
Vinaigrette (maximum 1 tablespoon per day; toss salads)
Whole-grain breads, crackers, and breakfast cereals
or until the first follow-up visit thereafter.
I performed a laryngeal examination with each office visit, as is the routine for management of LPR.
The reflux finding score (RFS)20 was calculated for
each patient for each visit; however, for this study,
it was not blinded, as it was anticipated that there
would be no significant change in the RFS. We have
previously reported that the laryngeal findings of
LPR do not usually change as quickly as the symptoms.19 (I never expected the degree of improvement
in the RFS that was found.) For statistical analysis
I used Students’ t-test for the pre-diet and post-diet
RSI and RFS data.
Institutional Review Board approval was not
sought for this study, as it was deemed unnecessary
because 1) the strict, low-acid, induction reflux diet
carried no risk of harm; 2) the diet was a logical extension of the traditional antireflux diet; 3) alternative therapeutic alternatives were neither denied nor
precluded; and 4) there was no risk of violation of
patient confidentiality, as no one but the author had
access to the study data.
RESULTS
There were 12 male and 8 female subjects with a
mean age of 54.3 years (range, 24 to 72 years). All
of the study subjects claimed complete compliance
with the prescribed diet. Nineteen of the 20 subjects
(95%) improved on the low-acid diet, and 1 got
worse. Three subjects became completely asymptomatic, and another went from an initial RSI score
of 28 to a post-diet RSI score of 4. The mean prediet RSI score was 14.9, and the mean post-diet RSI
score was 8.6 (p = 0.020); the mean RSI improvement was 6.3. The mean pre-diet RFS was 12.0, and
the mean post-diet RFS was 8.3 (p < 0.001). The
data are shown in Table 3.
DISCUSSION
In 1981, I was emergently consulted to see a patient with airway obstruction. Portable endoscope in
hand, I rushed to the hospital to see a stridulous patient. She calmly pointed to her throat and gasped,
“Can’t breathe…acid reflux.” After a quick bedside
endoscopy, I took her to the operating room and removed the two largest obstructing vocal process
granulomas that I have ever seen before or since.
The patient was placed on a postoperative regimen
of high-dose cimetidine, head-of-bed elevation, and
a restricted diet: no fried food, no coffee, no tomatoes, onions, garlic, cheese, chocolate, or mints, as
well as no late eating. Under that treatment, the patient got well. She was my introduction to LPR.
In the 30 years since, reflux medications have
evolved, and now many patients with LPR are started on “maximal antireflux treatment” consisting of
twice-daily PPIs (before breakfast and before the
evening meal) and an H2-receptor antagonist at bedtime.21 Although this regimen results in better acid
suppression than did previous medical therapy, there
is still a significant rate of medical treatment failure
(10% to 17%).22,23 It is presumed that PPI failure is
due to a “bioavailability” problem (ie, poor drug absorption).22
More than a decade ago, we recognized that carbonated beverages, particularly caffeinated cola
drinks, were a major risk factor for LPR. Indeed, excessive consumption of carbonated beverages was
the single most commonly identified cause of medical treatment failure among our patients with LPR.
Koufman, Low-Acid Diet for Laryngopharyngeal Reflux
69
TABLE 3. RESULTS
Subject
Age
(y)
1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
Mean
29
70
72
53
61
41
62
57
82
33
65
52
63
60
60
59
52
24
39
52
54.3
Sex
F
F
F
M
M
M
M
M
F
M
M
F
M
M
F
M
F
M
M
F
Reflux Symptom Index Score
Before
After
Diet
Diet
Change
28
4
10
10
29
18
20
1
2
12
27
21
13
7
7
19
21
22
15
12
14.9
On the basis of clinical experience, we also limited
our patients’ intake of citrus fruits and hot (pepper)
sauces. Other than these few specifics, the antireflux
diet has not changed much over the years — that is,
until recently. In 2008, we began measuring the pH
of common foods and beverages, and as a consequence of finding acid in almost everything we tested, we began to limit the acid intakes of our patients
with LPR, with surprisingly good results.
The clinical results reported herein are particularly striking and significant because “maximum antireflux therapy” was failing in the study patients. In
the months since this paper was presented, many additional patients with LPR have been treated with a
low-fat, low-acid diet as the cornerstone of therapy,
with or without adjunctive antireflux medications.
The induction reflux diet is still recommended for
the first 2 to 4 weeks with a gradual reintroduction
of some fatty foods and other historically “refluxogenic” foods. Cheese, eggs, meats, sauces, and condiments are allowed in moderation, but the key elements of the maintenance reflux diet are that it remains relatively low in acids and low in fat.
With fatty foods in particular, we teach patients
moderation, and to use tasty fats as flavorings, not
as main ingredients. We also introduce the concept
of pH balancing. The idea is that acidic foods may
be combined with nonacidic foods. Strawberries,
for example, which are not allowed to be eaten by
4
3
0
8
35
15
8
0
0
10
16
12
7
4
3
7
16
9
8
7
8.6
24
1
10
2
–6
3
12
1
2
2
11
9
6
3
4
12
5
13
7
5
6.3
Reflux Finding Score
Before
After
Diet
Diet
13
12
10
10
12
13
11
10
14
14
16
14
9
9
12
11
14
12
12
12
12.0
8
5
0
9
12
10
9
4
9
11
11
9
5
9
11
10
11
11
8
4
8.3
themselves on the reflux diet, are permitted when
added to breakfast cereal with milk, preferably lowfat milk, which has a high pH. In other words, the
cereal and milk buffer the acidic fruit; ie, they pHbalance the dish.
It is important to recognize that these ideas and
their practical applications in clinical practice have
evolved over a period of many years. By reporting a
series of worst-case, PPI-resistant patients who had
successful outcomes with a strict low-acid diet, it is
my hope to stimulate interdisciplinary research in
the areas of reflux, nutrition, and the American diet.
Indeed, the contemporary American diet appears to
be making Americans sick.
SCIENTIFIC BASIS FOR DIETARY ACID RESTRICTION
IN REFLUX MANAGEMENT
Despite the popular use of the term “acid reflux”
among the lay public, most of the clinical manifestations of LPR and GERD are due to pepsin.1-3,5,7-9,11-17
Pepsin is responsible for tissue injury and inflammation, and the confusion stems from the fact that
pepsin requires acid activation.1,14 The pepsin activity profile (see Figure), the cell biology of LPR, and
clinical experience with pharyngeal pH monitoring
all suggest that the threshold of the larynx for peptic
injury is far less than that of the esophagus.1,8,11-17
Surprisingly little acid is needed for peptic activation, and pepsin remains mildly proteolytic up to pH
6.5.14 In addition, tissue-bound pepsin can be acti-
70
Koufman, Low-Acid Diet for Laryngopharyngeal Reflux
vated by hydrogen ions from any (including a dietary) source.14
It appears that the key to the development of clinical laryngeal disease is the presence of tissue-bound
pepsin,8,9 which causes depletion of protective cell
proteins such as carbonic anhydrase, E-cadherin,
and the stress (“heat-shock”) proteins.8,9,12,13 Johnston et al8 demonstrated (in vitro and in vivo) that
peptic laryngeal damage occurs at pH 5. It was basic science that led us to consider the possibility that
the contemporary reflux epidemic might be related
to the contemporary American diet.
INCREASING PREVALENCE OF REFLUX DISEASE AND
REFLUX-RELATED ESOPHAGEAL CANCER
The prevalence of acid reflux disease (GERD
and LPR) has increased dramatically in our lifetimes.24,25 Using a Poisson model and an analysis of
17 prevalence studies, El-Serag24 showed that since
1976, the mean rate of increase of GERD has been a
staggering 4% per year (p < 0.0001).
Altman et al25 reported that office visits to otolaryngologists for LPR increased almost 500% from
1990 to 2001. Those authors hypothesized that the
increase was due to obesity and a greater awareness
of LPR by otolaryngologists25; however, reportedly,
only 27% of the study patients were counseled about
an antireflux diet.25
An even more ominous trend is the skyrocketing
increase in the prevalence of esophageal cancer in
the United States.26-30 The US National Cancer Institute data from 2005 reveal that esophageal cancer
had increased 600% since 1975 (from 4 to 23 cases
per million).26 During this same period, its mortality
rate increased sevenfold, despite increased esophageal surveillance26,27; the histopathology has been
trending toward more deadly, poorly differentiated
adenocarcinomas.27,28
In addition, the prevalence of Barrett’s esophagus (a reflux-related precursor to esophageal cancer) is also very high.27-30 Reavis et al29 reported
that patients with hoarseness, sore throat, and chronic cough (LPR symptoms) had Barrett’s esophagus
just as frequently (7% to 10%) as did patients with
GERD and heartburn. Thus, routine esophageal
screening for both LPR and GERD was (and still is)
recommended.29,30
INCREASED PREVALENCE OF REFLUX IN YOUNG
PATIENTS
In 2010, we estimated the prevalence of reflux
(GERD and LPR) in the United States by interviewing 656 adult US citizens while they were waiting
in line to purchase discount theater tickets in Times
Square in New York City. (This specific location
appeared to provide us with a reasonable approximation of a national sample.) The interviews were
carefully conducted to elicit all reflux symptoms
and medications, both over-the-counter and physician-prescribed. Respondents were considered to
have a tendency to reflux if they had multiple reflux symptoms and/or took reflux medications. For
the purposes of this survey, respondents with only
one symptom, such as hoarseness or cough, were
not considered to have reflux, as one symptom may
have many different causes.
The data revealed that an astonishing 40% (262
of 656) of the study group had reflux disease, with
22% (144 of 656) having classic GERD and another
18% (118 of 656) having LPR. There were no statistical differences between age groups, genders, and
regions of the country. The most striking and unanticipated result was that 37% of the 21- to 30-yearold age group had reflux.
In the past, reflux was primarily a disease of overweight, middle-aged people. Now, we are finding
that many of our reflux patients are neither old nor
obese.10 This trend toward younger and younger patients with more and more severe reflux has been
noted by other experienced clinicians (J. Hunter and
R. Sataloff, personal communications, 2011).
CHANGES IN AMERICAN DIET IN PAST FIFTY YEARS
Coincident with the reflux epidemic, the American diet has changed dramatically.31-36 Since the
1960s, there have been four parallel unhealthy dietary trends: 1) increased saturated fat; 2) increased
high-fructose corn syrup; 3) increased exposure to
organic pollutants (eg, DDT, PCBs, dioxins); and
4) increased acidity.33-35 The last of these trends —
increased dietary acid — may hold the key to understanding the contemporary reflux epidemic and
the dramatic increases in Barrett’s esophagus and
esophageal cancer.29,33,34
In 1973, after an outbreak of food poisoning
(botulism), the US Congress enacted Title 21, mandating that the US Food and Drug Administration
(FDA) ensure the safety of processed food crossing state lines by establishing “Good Manufacturing Practices.”33,34 How was this accomplished?
Through acidification of bottled and canned foods,
which was intended to prevent bacterial growth and
prolong shelf life. For two generations, the FDA has
never wavered from this path, apparently without
ever considering the possibility that the acidification of America’s food supply might have potential
adverse health consequences. From the 1979 Title
21 Act34:
Acidified foods should be so manufactured, processed,
and packaged that a finished equilibrium pH value of 4.6
Koufman, Low-Acid Diet for Laryngopharyngeal Reflux
or lower is achieved. If the finished equilibrium pH is
4.0 or below, then the measurement of acidity of the final
product may be made by any suitable method. [April 1,
2002; US Government Printing Office, 21CFR114.80]
In other words, the FDA actually encourages food
manufacturers to reduce the pH of their products to
less than 4.0, the same pH level as stomach acid.1 In
addition to acetic, ascorbic, citric, and hydrochloric
acids as food additives, the FDA allows more than
300 other chemicals that are “generally regarded as
safe” (GRAS).33-36 Many of these GRAS food additives were approved in the 1970s without the benefit
of contemporary methods of scientific testing and
analysis.34,36
Furthermore, in 1997, the FDA was directed to
provide specific criteria for food manufacturers to
report their use of GRAS additives in their products; inexplicably, as of this writing, those criteria
have still not been established.36 Thus, with regard
to food additives, the food industry remains completely self-regulated.33,36
In 2010, the US Government Accountability Office, a bipartisan group of scientists, published a
scathing report on the FDA’s lack of oversight of
food manufacturing.36 In particular, they were critical of the FDA’s negligence in failing to monitor
GRAS food additives, as referenced above.33-35 In
searching the literature, it appears that neither the
FDA nor the scientific community have examined
the acidity question. No one, it appears, has considered the possibility that the acidification of the
nation’s food might have potentially adverse health
consequences; today, almost all food that is bottled
or canned is below pH 4.33
It is interesting to note that the Amish, who grow
and consume their own organic food, have aerodigestive tract cancer rates (eg, larynx, pharynx,
esophagus) that are only 37% the rates of controls.37
In attempting to explain those findings, the authors
emphasized that the Amish do not drink alcohol or
smoke tobacco, but they did not discuss or consider
the possible health benefits of a diet devoid of acids
and other food additives.37
DIETARY ACID AS POSSIBLE MISSING LINK IN
REFLUX EPIDEMIC
Why are reflux disease and esophageal cancer
epidemic? Many years ago, when esophageal can-
71
cer was relatively uncommon, reflux patients usually presented in middle age. Today, we are seeing
comparable disease in patients in their twenties (J.
Hunter and R. Sataloff, personal communications,
2011). Overacidity in the diet may be the missing
link that explains the reflux epidemic, as well as increasing rates of Barrett’s esophagus and esophageal cancer.
At present, even baby food is acidified. We tested
the acidity of an “organic” banana baby food and
found it to be pH 4.3; normally, banana is pH 5.7.
The label of the so-called “organic” banana product revealed that it had had both ascorbic and citric
acids added. Indeed, almost all bottled and canned
foods and beverages contain ascorbic acid and/or
citric acid; sometimes the packaging is less obvious
and may just read “vitamin C–enriched” or “vitamin
C–enhanced.”33
Knowing what we now know about the cell biology of reflux, the stability and activity of pepsin,
and the contemporary American diet, it is reasonable to postulate that the acidification of America’s
food supply may be responsible for the reflux epidemic. Diet may be the primary factor in the prevalence, mechanisms, manifestations (including neoplasia), and outcomes of reflux disease, particularly as it affects the laryngopharynx and esophagus.
Further research is needed to investigate this question. Until now, it appears that fundamental nutritional issues related to how food has been preserved
for the past two generations may have been overlooked. Dietary acid may turn out to have serious
adverse health effects on the general population,
and how food is preserved may soon become a perplexing public health conundrum. In the meanwhile,
it seems likely that patients with LPR will benefit
from a low-acid diet.
CONCLUSIONS
A strict low-acid (“acid-free”) diet appears to be
beneficial for patients with pH-documented LPR.
In this study, the diet was shown to improve both
the symptoms and the laryngeal findings of patients
with recalcitrant (PPI-resistant) LPR. Also raised by
this study are broader public health policy issues related to FDA-mandated acidification of manufactured foods and beverages.
REFERENCES
1. Koufman JA. The otolaryngologic manifestations of gastroesophageal reflux disease (GERD): a clinical investigation
of 225 patients using ambulatory 24-hour pH monitoring and
an experimental investigation of the role of acid and pepsin in
the development of laryngeal injury. Laryngoscope 1991;101
(suppl 53):1-78.
2. Koufman JA. Perspective on laryngopharyngeal reflux:
from silence to omnipresence. In: Branski R, Sulica L, eds.
Classics in voice and laryngology. San Diego, Calif: Plural Publishing, 2009:179-266.
3. Koufman JA, Wiener GJ, Wu WC, Castell DO. Reflux
72
Koufman, Low-Acid Diet for Laryngopharyngeal Reflux
laryngitis and its sequelae: the diagnostic role of 24-hour pH
monitoring. J Voice 1988;2:78-9.
4. Postma GN, Tomek MS, Belafsky PC, Koufman JA.
Esophageal motor function in laryngopharyngeal reflux is superior to that of classic gastroesophageal reflux disease. Ann Otol
Rhinol Laryngol 2001;110:1114-6.
5. Axford SE, Sharp S, Ross PE, et al. Cell biology of laryngeal epithelial defenses in health and disease: preliminary studies. Ann Otol Rhinol Laryngol 2001;110:1099-108.
6. Amin MR, Postma GN, Johnson P, Digges N, Koufman
JA. Proton pump inhibitor resistance in the treatment of laryngopharyngeal reflux. Otolaryngol Head Neck Surg 2001;125:3748.
7. Koufman JA, Belafsky PC, Bach KK, Daniel E, Postma
GN. Prevalence of esophagitis in patients with pH-documented
laryngopharyngeal reflux. Laryngoscope 2002;112:1606-9.
8. Johnston N, Bulmer D, Gill GA, et al. Cell biology of
laryngeal epithelial defenses in health and disease: further studies. Ann Otol Rhinol Laryngol 2003;112:481-91.
9. Johnston N, Knight J, Dettmar PW, Lively MO, Koufman J. Pepsin and carbonic anhydrase isoenzyme III as diagnostic markers for laryngopharyngeal reflux disease. Laryngoscope
2004;114:2129-34.
10. Halum SL, Postma GN, Johnston C, Belafsky PC, Koufman JA. Patients with isolated laryngopharyngeal reflux are not
obese. Laryngoscope 2005;115:1042-5.
11. Knight J, Lively MO, Johnston N, Dettmar PW, Koufman JA. Sensitive pepsin immunoassay for detection of laryngopharyngeal reflux. Laryngoscope 2005;115:1473-8.
12. Gill GA, Johnston N, Buda A, et al. Laryngeal epithelial
defenses against laryngopharyngeal reflux: investigations of Ecadherin, carbonic anhydrase isoenzyme III, and pepsin. Ann
Otol Rhinol Laryngol 2005;114:913-21.
13. Johnston N, Dettmar PW, Lively MO, et al. Effect of pepsin on laryngeal stress protein (Sep70, Sep53, and Hsp70) response: role in laryngopharyngeal reflux disease. Ann Otol Rhinol Laryngol 2006;115:47-58.
14. Johnston N, Dettmar PW, Bishwokarma B, Lively MO,
Koufman JA. Activity/stability of human pepsin: implications
for reflux attributed laryngeal disease. Laryngoscope 2007;117:
1036-9.
15. Samuels TL, Johnston N. Pepsin as a marker of extraesophageal reflux. Ann Otol Rhinol Laryngol 2010;119:203-8.
16. Lillemoe KD, Johnson LF, Harmon JW. Role of the components of the gastroduodenal contents in experimental acid
esophagitis. Surgery 1982;92:276-84.
17. Johnson LF, Harmon JW. Experimental esophagitis in a
rabbit model. Clinical relevance. J Clin Gastroenterol 1986;8
(suppl 1):26-44.
18. Belafsky PC, Postma GN, Koufman JA. Validity and reliability of the reflux symptom index (RSI). J Voice 2002;16:2747.
19. Belafsky PC, Postma GN, Koufman JA. Laryngopharyngeal reflux symptoms improve before changes in physical findings. Laryngoscope 2001;111:979-81.
20. Belafsky PC, Postma GN, Koufman JA. The validity
and reliability of the reflux finding score (RFS). Laryngoscope
2001;111:1313-7.
21. Koufman JA, Aviv JE, Casiano RR, Shaw GY. Laryngopharyngeal reflux: position statement of the Committee on
Speech, Voice, and Swallowing Disorders of the American
Academy of Otolaryngology–Head and Neck Surgery. Otolaryngol Head Neck Surg 2002;127:32-5.
22. Amin MR, Postma GN, Johnson P, Digges N, Koufman
JA. Proton pump inhibitor resistance in the treatment of laryngopharyngeal reflux. Otolaryngol Head Neck Surg 2001;125:3748.
23. El-Serag H, Becher A, Jones R. Systematic review: persistent reflux symptoms on proton pump inhibitor therapy in
primary care and community studies. Aliment Pharmacol Ther
2010;32:720-37.
24. El-Serag HB. Time trends of gastroesophageal reflux disease: a systematic review. Clin Gastroenterol Hepatol 2007;5:
17-26.
25. Altman KW, Stephens RM, Lyttle CS, Weiss KB. Changing impact of gastroesophageal reflux in medical and otolaryngology practice. Laryngoscope 2005;115:1145-53.
26. Pohl H, Welch HG. The role of overdiagnosis and reclassification in the marked increase of esophageal adenocarcinoma
incidence. J Natl Cancer Inst 2005;97:142-6.
27. Lund O, Hasenkam JM, Aagaard MT, Kimose HH. Timerelated changes in characteristics of prognostic significance in
carcinomas of the oesophagus and cardia. Br J Surg 1989;76:
1301-7.
28. Conio M, Blanchi S, Lapertosa G, et al. Long-term endoscopic surveillance of patients with Barrett’s esophagus. Incidence of dysplasia and adenocarcinoma: a prospective study.
Am J Gastroenterol 2003;98:1931-9.
29. Reavis KM, Morris CD, Gopal DV, Hunter JG, Jobe BA.
Laryngopharyngeal reflux symptoms better predict the presence
of esophageal adenocarcinoma than typical gastroesophageal
reflux symptoms. Ann Surg 2004;239:849-58.
30. Amin MR, Postma GN, Setzen M, Koufman JA. Transnasal esophagoscopy: a position statement from the American
Broncho-Esophagological Association. Otolaryngol Head Neck
Surg 2008;138:411-4. [Erratum in Otolaryngol Head Neck Surg
2009;140:280.]
31. Bellis M. Introduction to pop: the history of soft drinks
timeline. About.com (http://inventors.about.com/od/sstartinventions/a/soft_drink.htm). Accessed March 2010.
32. Lobbying 2009: American Beverage Association. Center for Responsive Politics. (http://www.opensecrets.org/lobby/
clientlbs.php?year=2009&lname=American+Beverage+Assn
&id). Accessed March 2010.
33. Koufman JA, Stern JC, Bauer MM. Dropping acid: the
reflux diet cookbook and cure. Minneapolis, Minn: Reflux
Cookbooks, 2010.
34. Acidified foods. Code of Federal Regulations — Title 21
— Food and Drugs Chapter I, Department of Health and Human
Services Subchapter B — Food for Human Consumption Part
114. United States Food and Drug Administration. Arlington,
Va: Washington Business Information, 2010.
35. Generally recognized as safe food additives: FDA database of selected GRAS substances. United States Food and
Drug Administration. Springfield, Va: National Technical Information Service, 2009.
36. Food safety: FDA should strengthen its oversight of
food ingredients determined to be generally recognized as safe
(GRAS). GAO-10-246, United States Government Accountability Office, February 3, 2010.
37. Westman JA, Ferketich AK, Kauffman RM, et al. Low
cancer incidence rates in Ohio Amish. Cancer Causes Control
2010;21:69-75.
Cooling the “Oven”: A Temperature Study of Air and Glottal
Tissue During Laser Surgery in an Ex Vivo Calf Larynx Model
James A. Burns, MD; Gerardo Lopez-Guerra, MD; James T. Heaton, PhD;
James B. Kobler, PhD; Julie Kraas; Steven M. Zeitels, MD
Objectives: Endoscopic microlaryngeal laser surgery performed with general anesthesia through a laryngoscope speculum generates heat that accumulates at the distal lumen, creating an “oven” effect and potentially causing bystander thermal damage to nontarget tissue such as the contralateral vocal fold. We report the effects of cooling on air and tissue temperatures that occurred during simulated laryngeal laser surgery with KTP and thulium lasers in an ex vivo calf model.
Methods: Ten fresh excised calf larynges were studied at room temperature. Laser energy was applied to one vocal fold
for 2 minutes, with or without cooling, while temperatures were monitored with sensors placed within the glottal lumen
or inserted superficially into the contralateral vocal fold. A pulsed KTP laser (525 mJ) was used for 5 larynges, and a thulium laser (7 W, continuous) was used for the other 5 larynges.
Results: Heating was slightly greater for the KTP laser than for the thulium laser with use of these parameters. The lumen temperatures for both lasers increased an average of 13.2°C without cooling, but only 6.7°C with cooling (p < 0.05).
The contralateral vocal fold (subepithelial space) temperature increased an average of 6.8°C without cooling, but only
4.2°C with cooling (p > 0.05).
Conclusions: Cooling with room-temperature air during laryngeal laser surgery reduces luminal air and contralateral vocal fold temperatures. This effect is believed to be due to elimination of the plume of steam and smoke that significantly
heats surrounding structures.
Key Words: cooling, laser, microlaryngoscopy, thermal damage, vocal cord, vocal fold.
ately adjacent to the primary target of the laser. Excessive heat can lead to epithelial sloughing, direct
damage to the underlying superficial lamina propria,
and excessive scarring and fibrosis.16,17
INTRODUCTION
Endoscopic laryngeal laser surgery has gained
broad acceptance for the treatment of various benign and malignant disorders. Since the initial clinical use of the carbon dioxide laser by Strong and
Jako1,2 and Vaughan,3 the variety of available lasers
has expanded to provide both cutting-ablating properties4,5 and selective photoangiolysis.6-9 All lasers
commonly used in endolaryngeal surgery generate
heat, and the thermal effects of lasers on laryngeal
tissue are well documented both in ex vivo animal
models4,10-14 and in human clinical series.3,5,7,8,15 Although the thermal effect of lasers can be favorable
(such as in coagulation of small blood vessels for
optimal hemostasis during extensive endolaryngeal
resection5), the heat generated by lasers can also be
unfavorable. Collateral thermal damage should be
minimized during endoscopic laser procedures, especially when the layered microstructure of the vocal folds either is the primary target or is immedi-
Endoscopic microlaryngeal laser surgery performed with general anesthesia through a rigid laryngoscope generates heat that accumulates within
the confined space of the distal lumen, creating an
“oven” effect. Careful intraoperative observation
can reveal evidence of tissue singeing unrelated to
direct application of laser energy to endolaryngeal
structures. Thus, it appears that “bystander” thermal
damage can occur separate from the target tissue,
such as on the contralateral vocal fold (Fig 1). Although previous work has shown that precooling diminishes thermal effects at the target site,13 no prior
study has examined the effects of cooling on luminal air temperature at the distal laryngoscope or on
temperature within the superficial lamina propria
of the contralateral vocal fold. Cooling tissue with
From the Department of Surgery, Harvard Medical School, and the Center for Laryngeal Surgery and Voice Rehabilitation, Massachusetts General Hospital, Boston, Massachusetts. This work was supported in part by the Institute of Laryngology and Voice Restoration
and the Eugene B. Casey Foundation. Dr Zeitels has an equity interest in Endocraft LLC, which makes the glottiscope that was used
in the study.
Presented at the meeting of the American Broncho-Esophagological Association, Las Vegas, Nevada, April 28-29, 2010.
Correspondence: James A. Burns, MD, Center for Laryngeal Surgery and Voice Rehabilitation, One Bowdoin Square, 11th Floor,
Boston, MA 02114.
73
74
Burns et al, Cooling During Glottal Laser Surgery
Fig 1. Endoscopic surgery with use of KTP laser to treat
left glottal cancer. Disease is unilateral, with laser being directed at left-sided lesion (A). During treatment, debris and
singed tissue accumulate on contralateral vocal fold, as seen
on medial edge (B) and on undersurface (C) of right vocal
fold. Right vocal fold is being retracted laterally to show
undersurface.
A
C
B
pressurized room air offers a less expensive alternative to cooling devices that generate hypothermic
airflow, because most standard operating rooms are
already equipped with pressurized air. This article
reports the cooling effects of directed pressurized
room-temperature air on lumen and glottal tissue
temperatures during simulated laryngeal laser surgery with KTP (potassium-titanyl-phosphate) and
thulium lasers in an ex vivo calf model.
MATERIALS AND METHODS
Fresh calf larynges were obtained from a meatpacker and maintained on ice until being warmed
to room temperature at the time of the experiment.
Standard intraoperative conditions typical of endoscopic microlaryngeal laser surgery as performed
through a rigid laryngoscope were replicated as far
as possible in the ex vivo benchtop model (Fig 2).
These conditions included placement of a standard
cuffed endotracheal tube to block the distal egress of
heat and the use of suctioning during application of
laser energy. A Universal Modular Glottiscope (Endocraft LLC, Providence, Rhode Island) was used
to obtain exposure of the glottal surface that was the
target for laser application.
Endoscopic laryngeal laser surgery was simulated
in 10 excised calf larynges with two commonly used
lasers: the 2.01-μm thulium laser (Revolix Jr, LISA
Laser, Katlenburg-Lindau, Germany; 400-μm fiber) and the 532-nm KTP laser (Aura XP with StarPulse, Laserscope Inc, San Jose, California; 400-μm
fiber). The thulium laser (7 W continuous power)
was used to make a controlled cut on the mid-musculomembranous and paraglottic regions of 5 calf
larynges. Laser energy was evenly applied over one
vocal fold surface while temperature measurements
were taken from the glottal lumen and subepithelial
tissue layer of the contralateral vocal fold. Cutting
was performed for 2 minutes without application of
a cooling airflow, and 2 minutes with cooling. The
KTP laser (525 mJ, 15-ms pulse width, 2 pulses per
second) was likewise used to cut and ablate one vocal fold for 2 minutes in 5 calf larynges, with and
without the cooling airflow. The excised calf larynges did not contain perfusing tissue, so there was no
selective uptake of laser energy from this photoangiolytic laser; rather, heat was generated by the nonspecific application of laser energy to the tissue.
Temperature measurements were obtained with
a 1.0-mm thermistor (Thermometrics, Edison, New
Burns et al, Cooling During Glottal Laser Surgery
75
Fig 2. Procedural setup of ex vivo
calf model simulating endoscopic
microlaryngeal laser surgery. Inset) Typical laser wound on left
vocal fold, along with thermometer probe and thermistor.
Jersey) and a contact thermometer (Omega dual input HH506R, Omega Engineering Inc, Stamford,
Connecticut) that were placed within the immediate subepithelial space of the vocal fold contralateral to the laser application and in the glottal lumen,
respectively. The thermometer within the glottal
lumen was positioned to measure luminal air temperature near the tip of the glottiscope. Temperature
measurements from within the superficial lamina
propria of the contralateral vocal fold were obtained
by placing a thermistor into this position through a
lateral puncture with an 18-gauge carrier needle and
confirming its subepithelial placement visually.
with cooling. For each 2-minute laser application,
the average temperature of the hottest 5 consecutive
seconds (Tavg) was calculated by custom software
written in Matlab (MathWorks Inc, Natick, Massachusetts). Cooling was applied by directing pressurized room-temperature air through a second catheter
affixed parallel to the laser catheter. The lowest rate
of airflow (2 L/min) that would eliminate the visible plume of steam and smoke during lasering was
chosen (Fig 3). The Tavg values were compared for
the cooled versus noncooled conditions collapsed
across laser type with 1-tailed Student’s t-tests (alpha level of less than 0.05 for significance).
For each calf larynx and laser application, temperature measurements during lasering without
cooling were obtained first, followed by lasering
RESULTS
A
The heating effects were very similar for the KTP
B
Fig 3. Plume of steam obscures optimal view of calf vocal fold during simulated laser surgery using thulium laser without
cooling (A). Effective cooling with application of pressurized room-temperature air through second lumen next to laser
catheter creates clear view of vocal folds with no visible steam plume (B). Blue thermistor tip used to measure ambient
air temperature at glottiscope tip is seen in upper left corner of each image.
76
Burns et al, Cooling During Glottal Laser Surgery
peratures reached 29.0°C (KTP) and 24.9°C (thulium), which were decreased to about 23°C (slightly
higher than room temperature) for both lasers with
cooling.
DISCUSSION
Fig 4. Graphic representation of luminal air (in lumen) and
contralateral vocal fold (in tissue) temperature measurements obtained for each laser with and without cooling.
and thulium lasers with use of these parameters. Because both lasers showed the same trend in terms of
their effect on luminal air and glottal tissue temperatures, as well as their response to cooling, the results
for these two lasers were averaged. Temperature recordings with and without cooling are reported for
measurements made within the luminal air at the tip
of the glottiscope and within the immediate subepithelial (superficial lamina propria) space of the contralateral vocal fold. Figure 4 summarizes the temperature measurements obtained for each laser with
and without cooling in the luminal air and contralateral vocal fold. Note that the ex vivo calf larynx
tissue began at room temperature (approximately
20°C).
Luminal Air. Without cooling, the luminal Tavg
for both lasers (10 specimens) at the tip of the glottiscope increased an average of 13.2°C. With cooling,
luminal Tavg was reduced nearly 50%, to only 6.7°C
(degrees of freedom, 9; p < 0.05). The peak luminal temperatures without cooling reached as high as
39.7°C (KTP) and 34.7°C (thulium), having started at about 20°C, or room temperature. Lower peak
temperatures for each laser were noted with cooling
(29.9°C for KTP and 31.8°C for thulium).
Contralateral Vocal Fold. Without cooling, the
contralateral vocal fold (subepithelial space) temperature for both lasers (10 specimens) increased an
average of 6.7°C. Debris in the form of charred bits
of tissue accumulated on the endolaryngeal tissues
during lasering without cooling. With cooling, the
average increase in the contralateral vocal fold was
slightly reduced to only 4.2°C, which was not significantly different than the noncooled condition (degrees of freedom, 8; p > 0.05). However, there was
no accumulation of debris on the endolaryngeal tissues with cooling. Peak contralateral vocal fold tem-
Prior studies evaluating the effect of cooling during laser surgery have focused on reducing thermal
injury at the target site.13,14 On the basis of results
obtained in those studies, we now routinely attach a
cooling device when cutting-ablating laryngeal tissue with the thulium laser. Since evidence suggests
that cooling the target tissue during pulsed KTP laser photoangiolysis can impede optimal vessel coagulation at standard power and pulse-width settings,14 cooling is not routinely utilized during these
procedures. Although this judicious use of cooling
has reduced thermal effects at the site of disease and
allowed for selective vessel coagulation, intraoperative and postoperative observations have occasionally revealed evidence of tissue singeing at distant
sites such as the contralateral vocal fold. Heated tissue debris produced during laser cutting and carried
in the steam and smoke plume can be seen deposited
throughout the endolaryngeal tissues. Additionally,
epithelial sloughing has been detected after operation, consistent with a thermal injury from contact
with elevated intraluminal air temperatures. Clearly,
the thermal effects from heat-trapping at the distal
glottiscope have the potential to impair the structure
and function of otherwise normal laryngeal tissue.
The results of this study help us to define the scope
of the problem in terms of the magnitude of the temperature elevation and to work toward a solution to
eliminate this problem.
Luminal air temperatures increased an average of
13°C above baseline after 2 minutes of standard lasering technique that included the usual suction devices employed to evacuate smoke and debris from
the treatment site. The average temperature of the
hottest 5 consecutive seconds (Tavg) was reported in
this study because of the highly variable temperature
readings that were obtained throughout the 2-minute
laser period. This variability seemed to be related to
the amount of visible smoke and debris, with higher temperature measurements recorded when more
smoke was present. Suctioning did not always immediately eliminate the smoke and debris, allowing
for accumulation of heat at the distal end of the glottiscope and creation of an “oven” effect as lasering
continued. This same phenomenon is occasionally
seen during operation when the suction catheter is
occluded from debris or from abutting tissue during retraction. Since certain common laryngeal laser procedures (both KTP and thulium) require more
Burns et al, Cooling During Glottal Laser Surgery
than 30 minutes of total laser time, the “oven” effect
is potentially magnified beyond the effect measured
in this study.
In this ex vivo model, the glottal tissue started at
room temperature (approximately 20°C) and the luminal air temperatures reached peak values approximately 19°C above baseline. Extrapolation of this
result to actual endoscopic laryngeal laser surgery,
with starting temperatures at 37°C (body temperature), predicts peak temperatures near 57°C. Temperatures in this range can cause irreversible structural changes ranging from epithelial damage and
local inflammation18,19 to protein denaturation.20
Heat dissipation from blood flow through perfusing
tissue is presumed to be negligible in this scenario,
as the accumulation of heat over time would overwhelm the tissue’s capacity to dissipate the heat.
Thus, results from the ex vivo calf larynx are expected to be similar to perfusing laryngeal tissue
models in this regard.
Models have been established that can be used to
predict the quantitative effect of raising air temperature on the potential for injury to the surrounding
tissue. For example, Leach et al19 used a guinea pig
skin model to demonstrate the degree of epithelial
damage from graded temperature exposures. Temperatures of 47°C for up to 6 minutes of exposure
time produced no change, 1-minute exposures of
50°C to 55°C caused epithelial sloughing, and just
10 to 20 seconds of exposure at 70°C to 80°C created a severe burn injury. Exposing the phonatory layered microstructure to these temperatures for even
short periods of time can possibly lead to epithelial
sloughing and web formation, especially if extensive lasering is occurring near the anterior commissure.
Cooling the “oven” within the luminal air at the
glottal surface by directing pressurized room-temperature air through a second lumen next to the laser
catheter reduced the temperature elevation by nearly
50%. In the clinical scenario of endoscopic laryngeal laser surgery, this could keep the peak temperatures below 47°C and thereby greatly reduce the
risk of thermal damage to collateral structures. Our
results also showed that subepithelial tissue temperatures within the contralateral vocal fold were reduced with cooling (although not statistically significant), although the average temperature increase
was much less than the increase observed in luminal
air temperature during lasering (6.7°C and 13.2°C,
respectively).
Effective cooling in this study required the continuous flow of air near the target tissue to keep debris and smoke from collecting at the distal glotti-
77
scope. Clinically, there is a natural tendency for heat
to accumulate in the area between the endotracheal
balloon and the glottiscope tip, because there is little circulation of air and no natural egress. Without
cooling, heat will eventually be vented through the
glottiscope and through the suction cannula, but not
before reaching peak temperatures of approximately 19°C above baseline. Evaporative cooling of tissue and convection that blows away the steam and
smoke plume are the presumed mechanisms of action of effective cooling of the “oven” that was demonstrated herein.
Utilizing the ex vivo calf vocal fold is an effective
and efficient way to obtain initial data regarding the
thermal effects of lasers and the response to cooling. Although there are obvious limitations to this
model, such as variations in soft tissue hydration,
temperature, and turgor in comparison to perfusing
tissue, the data are useful in calibrating optimal parameters for lasering and cooling laryngeal tissue.
The temperature elevations recorded within the luminal air and subepithelial space of the contralateral
vocal fold during this study support the concept that
thermal damage can occur unrelated to the primary treatment of disease. Further study in a perfusing animal model, such as the rat larynx model proposed by Mallur et al,21 or by direct measurements
of air and tissue temperatures during human surgery
would overcome the limitations of an ex vivo model
and perhaps yield further information that would directly improve surgical outcomes. Future investigations will seek to quantify the extent of tissue damage from lasering created by exposure to elevated
luminal air temperatures over various durations of
time.
CONCLUSIONS
1. Cooling with room-temperature air during laryngeal surgery with KTP and thulium lasers decreases the average rise in luminal air temperatures
by nearly 50% in this ex vivo model.
2. Laryngeal structures such as the contralateral vocal fold that are distant from the target tissue
are subject to temperature increases and can be effectively cooled with pressurized room-temperature
air.
3. Effective cooling is believed to be due to elimination of the steam and smoke plume that accumulates near the treatment site and heats surrounding
structures.
4. Thermal damage from indirect laser-generated
heat should be quantified in live-perfusing models
in future studies.
78
Burns et al, Cooling During Glottal Laser Surgery
REFERENCES
1. Strong MS, Jako GJ. Laser surgery in the larynx. Early
clinical experience with continuous CO2 laser. Ann Otol Rhinol
Laryngol 1972;81:791-8.
2. Strong MS. Laser management of premalignant lesions
of the larynx. Can J Otolaryngol 1974;3:560-3.
3. Vaughan CW. Transoral laryngeal surgery using the CO2
laser: laboratory experiments and clinical experience. Laryngoscope 1978;88:1399-420.
4. Shapshay SM. Laser applications in the trachea and bronchi: a comparative study of the soft tissue effects using contact
and noncontact delivery systems. Laryngoscope 1987;97(suppl
41):1-26.
5. Zeitels SM, Burns JA, Akst LM, Hillman RE, Broadhurst
MS, Anderson RR. Office-based and microlaryngeal applications of a fiber-based thulium laser. Ann Otol Rhinol Laryngol
2006;115:891-6.
6. McMillan K, Shapshay SM, McGilligan JA, Wang Z, Rebeiz EE. A 585-namometer pulsed dye laser treatment of laryngeal papillomas: preliminary report. Laryngoscope 1998;108:
968-72.
7. Franco RA Jr, Zeitels SM, Farinelli WA, Anderson RR.
585-nm Pulsed-dye laser treatment of glottal papillomatosis.
Ann Otol Rhinol Laryngol 2002;111:486-92.
8. Burns JA, Zeitels SM, Akst LM, Broadhurst MS, Hillman RE, Anderson RR. 523-nm Pulsed potassium-titanyl-phosphate laser treatment of laryngeal papillomatosis under general
anesthesia. Laryngoscope 2007;117:1500-4.
9. Broadhurst MS, Akst LM, Burns JA, et al. Effects of 532nm pulsed-KTP laser parameters on vessel ablation in the avian
chorioallantoic membrane: implications for vocal fold mucosa.
Laryngoscope 2007;117:220-5.
10. Rebeiz EE, Aretz HT, Shapshay SM, Pankratov MM. Application of pulsed and continuous wave 1.32 and 1.06 microns
wavelengths of the Nd:YAG laser in the canine tracheobronchial tree: a comparative study. Lasers Surg Med 1990;10:501-9.
11. April MM, Rebeiz EE, Aretz HT, Shapshay SM. Endo-
scopic holmium laser laryngotracheoplasty in animal models.
Ann Otol Rhinol Laryngol 1991;100:503-7.
12. Sullivan CA, Rader A, Abdul-Karim FW, Abbass H, Moher RM. Dose-related tissue effects of the CO2 and noncontact
Nd:YAG lasers in the canine glottis. Laryngoscope 1998;108:
1284-90.
13. Burns JA, Kobler JB, Heaton JT, Lopez-Guerra G, Anderson RR, Zeitels SM. Thermal damage during thulium laser
dissection of laryngeal soft tissue is reduced with air cooling: ex
vivo calf model study. Ann Otol Rhinol Laryngol 2007;116:8537.
14. Burns JA, Kobler JB, Heaton JT, Anderson RR, Zeitels
SM. Predicting clinical efficacy of photoangiolytic and cutting/
ablating lasers using the chick chorioallantoic membrane model: implications for endoscopic voice surgery. Laryngoscope
2008;118:1109-24.
15. Herdman RC, Charlton A, Hinton AE, Freemont AJ. An
in vitro comparison of the erbium:YAG laser and the carbon dioxide laser in laryngeal surgery. J Laryngol Otol 1993;107:90811.
16. Stern LS, Abramson AL, Grimes GW. Qualitative and
morphometric evaluation of vocal cord lesions produced by the
carbon dioxide laser. Laryngoscope 1980;90:792-808.
17. Meyers A. Complications of CO2 laser surgery of the larynx. Ann Otol Rhinol Laryngol 1981;90:132-4.
18. Barbet JP, Chauveau M, Labbé S, Lockhart A. Breathing
dry air causes acute epithelial damage and inflammation of the
guinea pig trachea. J Appl Physiol 1988;64:1851-7.
19. Leach EH, Peters RA, Rossiter RJ. Experimental thermal
burns, especially the moderate temperature burn. Exp Physiol
1943;32:67-86.
20. Clark JH. Denaturation changes in egg albumin with
urea, radiation, and heat. J Gen Physiol 1943;27:101-11.
21. Mallur PS, Amin MR, Saltman BE, Branski RC. A model
for 532-nanometer pulsed potassium titanyl phosphate (KTP)
laser-induced injury in the rat larynx. Laryngoscope 2009;119:
2008-13.
Quantity and Three-Dimensional Position of the Recurrent and
Superior Laryngeal Nerve Lower Motor Neurons in a Rat Model
Philip Weissbrod, MD; Michael J. Pitman, MD; Sansar Sharma, MD;
Aaron Bender; Steven D. Schaefer, MD
Objectives: We sought to elucidate the 3-dimensional position and quantify the lower motor neurons (LMNs) of the recurrent laryngeal nerve (RLN) and the superior laryngeal nerve (SLN) in a rat model. Quantification and mapping of these
neurons will enhance the usefulness of the rat model in the study of reinnervation following trauma to these nerves.
Methods: Female Sprague-Dawley rats underwent microsurgical transection of the RLN, the SLN, or both the RLN and
SLN or sham surgery. After transection, either Fluoro-Ruby (FR) or Fluoro-Gold (FG) was applied to the proximal nerve
stumps. The brain stems were harvested, sectioned, and examined for fluorolabeling. The LMNs were quantified, and
their 3-dimensional position within the nucleus ambiguus was mapped.
Results: Labeling of the RLN was consistent regardless of the labeling agent used. A mean of 243 LMNs was documented for the RLN. The SLN labeling with FR was consistent and showed a mean of 117 LMNs; however, FG proved to
be highly variable in labeling the SLN. The SLN LMNs lie rostral and ventral to those of the RLN. In the sham surgical
condition, FG was noted to contaminate adjacent tissues — in particular, in the region of the SLN.
Conclusions: Fluorolabeling is an effective tool to locate and quantify the LMNs of the RLN and SLN. The LMN positions and counts were consistent when FR was used in labeling of either the RLN or the SLN. Fluoro-Gold, however,
because of its tendency to contaminate surrounding structures, can only be used to label the RLN. Also, as previously
reported, the SLN LMNs lie rostral and ventral to those of the RLN. This information results in further clarification of a
rat model of RLN injury that may be used to investigate the effects of neurotrophic factors on RLN reinnervation.
Key Words: animal model, Fluoro-Gold, Fluoro-Ruby, laryngeal nerve, lower motor neuron.
INTRODUCTION
cles. Both of these considerations require resolution
at the laryngeal muscle and the laryngeal motor neuron in the brain stem. An animal model is critical
to understanding the cause of synkinesis and later
devising treatments to potentiate the proper regeneration of the RLN. Such a model would need to be
investigated by endoscopic, electromyographic, and
histologic techniques to elucidate the neurologic regenerative processes that occur at both the larynx
and the lower motor neurons (LMNs) in the nucleus ambiguus (NA). In the present study, we aimed
to update the rat model by elucidating the 3-dimensional organization of the LMNs of the RLN and the
SLN. We provide an accurate reference for future
experiments involving retrograde labeling via muscle injections and direct application of tracers to the
RLN after injury. These future experiments will enhance our understanding of the mechanism of synkinesis and exploration of treatment to restore proper
Iatrogenic injury to the laryngeal nerves is a relatively common complication of thyroid, cervical,
or cardiothoracic surgery. The recurrent laryngeal
nerve (RLN) and the superior laryngeal nerve (SLN)
serve as conduits of both motor information and sensory information to and from the larynx. The RLN
innervates both adductor and abductor muscles in
the larynx. After transection of the RLN, the aberrant regeneration of the axons results in synkinesis
that leads to vocal fold immobility.
Synkinesis of laryngeal muscles may be considered in two ways. In one, a single regenerating RLN
axon may innervate both adductor and abductor
muscles. Conversely, regenerating axons may inappropriately innervate the adductor or abductor muscle, with abductor axons innervating adductor muscles and adductor axons innervating abductor mus-
From the Department of Otolaryngology, New York Eye and Ear Infirmary, New York (Weissbrod, Pitman, Schaefer), and the Department of Cell Biology, New York Medical College, Valhalla (Sharma, Bender), New York. This study was performed in accordance with
the PHS Policy on Humane Care and Use of Laboratory Animals, the NIH Guide for the Care and Use of Laboratory Animals, and
the Animal Welfare Act (7 U.S.C. et seq.); the animal use protocol was approved by the Institutional Animal Care and Use Committee
(IACUC) of New York Medical College.
Presented at the meeting of the American Broncho-Esophagological Association, Las Vegas, Nevada, April 28-29, 2010.
Correspondence: Michael J. Pitman, MD, 310 E 14th St, 6th Floor, New York Eye and Ear Infirmary, New York, NY 10003.
79
80
Weissbrod et al, Recurrent & Superior Laryngeal Nerve Lower Motor Neurons
function via accurate reinnervation.
Previous studies have examined the NA in rats
by using retrograde labeling with variable results.1-6
The current study was intended to reexamine the location and quantity of LMNs within the NA, and
specifically the relationship between the RLN LMNs
and the SLN LMNs, through the use of more sophisticated labeling agents that enable complete visualization of neurons, dendrites, and axons. We chose
Fluoro-Ruby (FR; Invitrogen, Carlsbad, California)
and Fluoro-Gold (FG; Fluorochrome LLC, Denver,
Colorado) because they are easily processed and allow for clear visualization of labeled cells. Via retrograde transport, these chemicals avidly label LMNs
and their processes in a relatively short period of
time and remain in the cell bodies for many months
without transsynaptic labeling.7
METHODS
Experimental Animals. Twenty female Sprague
Dawley rats weighing 250 g were used in the present study. Humane care was provided for the animals, and all institutional and national guidelines
were observed. The animals underwent transection
of the right RLN only (4 animals), the right SLN
only (4 animals), or both the RLN and SLN (8 animals) or sham surgery (4 animals). After transection, the proximal nerve stumps were soaked in labeling agent consisting of 5% FG in 0.1 mol/L phosphate-buffered saline solution (PBS) or 5% FR in
PBS for a period of 5 minutes for retrograde labeling of the neurons in the brain stem. The transected
nerve end was isolated from the surrounding fascia
and smooth muscles. Gelfoam soaked with the fluorochrome was then packed around the transected
nerve end and left in place for 5 minutes. After this
procedure, the cut nerve was thoroughly washed
with PBS and the area was dried with cotton swabs
to minimize contamination of the dye in the surrounding tissue. This technique was an alternative
to placing the nerve end in a well filled with fluorochrome. In previous experiments, the well technique
appeared to result in more nerve end trauma, as well
as contamination of surrounding tissue.
The choice of these fluorochromes was based
upon our previous work. Fluoro-Gold may penetrate intact axons and label other neurons in the
brain stem. This situation was avoided by the isolation and washing of the nerve after exposure to the
fluorochrome. Fluoro-Ruby, on the other hand, has
a higher molecular weight, and its use in the central
nervous system of the rat caused no erroneous labeling.7 Fluoro-Gold and FR labeling was performed in
4 groups of animals. In one group of 4 animals, the
RLN was labeled with FG and the SLN with FR. In
a second group of 4, the RLN was labeled with FR
and the SLN with FG. In a third group (8 total), 4
animals had only the RLN labeled with FR and the
other 4 had only the SLN labeled with FR. Sham
surgery and labeling was performed on a fourth
group of 4 animals.
Surgical Procedures. Each animal was sedated
with isoflurane and then received an intramuscular injection of 70 mg/kg of ketamine hydrochloride
and 7 mg/kg xylazine hydrochloride to produce sufficient anesthesia. The anterior cervical region was
injected subcutaneously with 0.2 mL of 1% lidocaine hydrochloride with epinephrine (1:100,000).
Anesthesia was confirmed with a tail and foot pinch
before the surgical procedure commenced.
The animals were placed supine, and a vertical
midline incision was made extending to the sternum. The strap muscles were separated in the midline and retracted with an eyelid retractor. With an
operating microscope (Carl Zeiss AG, Oberkochen,
Germany), the RLN was identified in the tracheoesophageal groove and isolated from its surrounding structures. The SLN was identified as it coursed
horizontally to the cricothyroid muscle. Depending
on the experimental condition, the RLN, the SLN,
or both the RLN and SLN were sharply transected.
The RLN was transected at the level of the seventh
tracheal ring, and the SLN was transected just proximal to its entry into the larynx. After transection,
fluorochrome was applied to the proximal nerve
stump for a period of 5 minutes. In cases in which
dual labeling was performed, cotton was placed between the labeling sites to prevent contamination. In
all experiments, only the right RLN and right SLN
were transected, with the contralateral side serving
as a control. After nerve labeling, the area was blotted with cotton to remove excess tracer, and the incision was closed with 3-0 silk suture. To confirm
injury to the RLN, we performed transoral laryngoscopy with a 0° rigid nasal endoscope (Karl Storz,
Tuttlingen, Germany) to confirm absence of movement and vocal fold paralysis.
For sham surgical procedures, the above process
was performed, including identification and isolation of the appropriate nerves. Gelfoam soaked with
fluorolabeling agent was packed around the uncut
nerve at the site for a period of 5 minutes. The Gelfoam was removed, the area was blotted with cotton,
and the wound was closed in the above fashion.
On postoperative day 10, the animals were painlessly sacrificed by lethal inhalation of isoflurane.
Immediately thereafter, the brain stems were harvested and placed for 4 hours in 4% paraformaldehyde
Weissbrod et al, Recurrent & Superior Laryngeal Nerve Lower Motor Neurons
81
Fig 1. Retrograde-labeled laryngeal motor neurons (LMNs). A)
Photomicrograph shows Fluoro-Gold (FG)–labeled recurrent
laryngeal nerve (RLN) LMNs in nucleus ambiguus (NA). Photo was taken with 20× objective. Scale bar — 100 μm. B) Photomicrograph shows Fluoro-Ruby (FR)–labeled RLN LMNs
in NA. Scale bar — 100 μm. C) Superimposed image of FGlabeled RLN and FR-labeled superior laryngeal nerve (SLN).
Area imaged is caudal region of NA. Note more rostral (top of
image) and lateral (right of image) orientation of SLN LMNs.
Scale bar — 200 μm.
A
B
in 0.1 mol/L pH 7.2 PBS. The isolated brain stems
were then transferred into 30% sucrose in PBS and
kept at 4°C until the tissue sank to the bottom of the
container. The tissue was embedded in optimal cutting temperature medium (OCT). The brain stems
were sectioned with a cryostat (Leica Microsystems,
Wetzlar, Germany) into 40-μm-thick serial, longitudinal sections and mounted onto precoated slides.
After allowing the sections to dry overnight, we irrigated sections in sterile saline solution to dissolve
the OCT, mounted them with 1:1 PBS-glycerin, and
cover-slipped them. The slides were kept at 4°C in
a closed container to avoid light contamination until
examination via fluorescent microscopy.
Quantification of Fluorolabeled LMNs. The brain
stem sections were examined by light microscopy
(Axioskop, Zeiss) by a reviewer who was unaware
of the surgical procedure performed. The slides
were systematically examined to ensure that all labeled cells were appropriately counted. Only cells
that had visible cell bodies and contained the nucleus were counted. The counts were confirmed by 2
other viewers in a blinded fashion. In specimens labeled with 2 fluorochromes, FR and FG interference
C
and absorption filters (Zeiss) were used to view and
capture images. Double images were then digitally
superimposed.
After quantification of cells, images were processed to assess stereotactic LMN position. Individual sections were digitized, and labeled cells within each section were demarcated digitally into an
x- and y-coordinate system. The perimeter of each
section was outlined digitally and assigned x- and ycoordinates as well. The z-coordinate was assigned
on the basis of specimen number within the serially
cut sections. Each section was then aligned in the
x- and y-planes in order to be centered on the preceding specimen. The x- and y-coordinates for each
cell were then recorded on a spreadsheet. The coordinates were then plotted in a 3-dimensional graph
with Sigmaplot (Systat Software Inc, Chicago, Illinois).
RESULTS
The NA in the brain stem of the rat has an unusual
territorial domain. It has two subdivisions: an upper part and a lower part. The upper subdivision lies
caudal to the facial nucleus, and the majority of the
82
Weissbrod et al, Recurrent & Superior Laryngeal Nerve Lower Motor Neurons
A
B
C
D
Fig 2. NA distribution of FG-labeled RLN (white) and FR-labeled SLN (gray). Note rostral, lateral, and ventral
orientation of SLN LMNs as compared to more dorsal, caudal, and medial position of RLN LMNs. A,B) FGlabeled LMNs are brought to front of image. C,D) FR-labeled LMNs are brought to front of image. Dashed line
demarcates level of obex.
SLN LMNs are located in it. The RLN LMNs are located in the ventral and caudal subdivision. We have
followed the classification of Bieger and Hopkins6
to identify the subdivisions of the NA.
When FR or FG was used on the transected RLN,
in 3 different experimental settings (total of 12 animals), the average number of labeled LMNs in the
ventral and caudal region of the NA was 243 (Fig
1A,B). When the SLN was labeled with FR, the average number of labeled LMNs in the NA was 117.
In the middle region of the two subdivisions of the
NA, there were labeled LMNs from the SLN and
the RLN. In cases in which the SLN and RLN were
labeled with separate dyes, there was overlap of labeled LMNs in the middle location of the subdivisions of the NA (Figs 1C, 2, and 3); however, we did
not observe any neurons that were double-labeled
with the two different fluorochromes. When the SLN
was labeled with FG, the number of labeled LMNs
was very high (mean, 422; 4 samples; see Table).
A reasonable consistency in the number of LMNs
labeled with FR was evident. It was also evident that
labeling of the RLN with either FG or FR gave a
consistent number of labeled LMNs. A major discrepancy in the number of labeled LMNs from the
SLN when FG was used as a tracer can be attributed
to the capability of FG to avidly label the LMNs of
injured muscles surrounding the SLN. The findings
suggest that the RLN is composed of a larger number of axons from the NA than is the SLN. Additionally, the use of FR in the present experiments provided more consistent and accurate labeling than did
the use of FG.
Statistical analysis using a 2-tailed t-test for cells
labeled through the RLN (12 samples) shows the
Weissbrod et al, Recurrent & Superior Laryngeal Nerve Lower Motor Neurons
83
Fig 3. Three-dimensional representation of relationship between RLN LMNs (white) and
SLN LMNs (gray). SLN LMNs are lateral, rostral, and ventral as compared to RLN LMNs.
There is also small area of overlap in medial
region of NA.
mean as 242.58 (SEM, 4.9; p < 0.001). In the SLN
(8 samples), the mean number of labeled cells was
117 (SEM, 8.9; p < 0.001). The group in which FG
was used to label the SLN was not included in the
statistical analysis.
In the sham surgical condition, when the intact
RLN or SLN was smeared with FR or FG, no neuNUMBERS OF LABELED NEURONS IN
NUCLEUS AMBIGUUS
Group
Fluorochrome
Tracer
RLN-Labeled
LMNs
SLN-Labeled
LMNs
A (n = 4)
Fluoro-Ruby
230
240
260
266
Mean = 249
120
129
130
90
Mean = 117
C (n = 8)
Fluoro-Gold
Fluoro-Ruby
243
246
239
236
Mean = 241
238
243
244
267
Mean = 248
2
2
B (n = 4)
311
430
529
420
Mean = 422
74
158
120
116
Mean = 117
Groups A and B represent one fluorochrome used to label recurrent
laryngeal nerve (RLN) and other fluorochrome used to label superior laryngeal nerve (SLN). Arrows point to dual-labeled animals.
Group C represents animals singly labeled with Fluoro-Ruby. LMNs
— lower motor neurons.
rons within the NA were labeled — a finding confirming that neither dye labeled intact axons in
the present experiments. Labeling with FG in the
transected nerves (both SLN and RLN) resulted in
scattered labeling of cells in the dorsal and peri-NA
regions. There was increased contamination noted
in FG-labeled SLN LMNs, which was likely due
to labeling of nerve endings in the dissected strap
muscles and nearby pharyngeal constrictor muscles.
No LMNs were labeled with FR when the nerves
were intact (sham experiments), likely because of
the higher molecular weight resulting in less-avid
uptake in surrounding muscle.
Two animals were excluded from the LMN count
studies after surgical transection and labeling; one
animal died in the perioperative period and the other
was not included because a number of sections were
destroyed during sectioning.
The LMNs of the SLN lie rostral, lateral, and ventral in relation to the more caudal, dorsal, and medial position of the RLN LMNs (Fig 2). The LMNs
of the RLN and SLN did share a region of overlap in
the more rostral portion of the RLN distribution. The
fact that there was no dual labeling of any LMNs
suggests that entirely separate LMN axons from the
NA traverse the RLN and SLN.
The majority of labeled neurons for the RLN and
SLN were confined within the NA. However, in 2
animals, a few labeled neurons were encountered in
the area corresponding to the contralateral NA. In 1
animal, 4 cells were noted, and 8 were noted in the
84
Weissbrod et al, Recurrent & Superior Laryngeal Nerve Lower Motor Neurons
other.
DISCUSSION
This study was designed as a complement to prior published work from our laboratory that details
techniques for studying reinnervation in rats and for
following animals after crush injury.8,9 These earlier studies focused on nerve injury via an aneurysm
clamp. In many of the studied conditions, the vocal
folds had purposeful movement after crush injury.
To ensure an experimental condition that reliably
and reproducibly creates synkinesis, we transected
the nerves instead of causing a crush injury. Nerve
transection leads to reliable and consistent nonfunctional synkinetic reinnervation of the larynx as evaluated by kinesiologic, histologic, and electromyographic parameters.10
The present work is a part of ongoing research on
rats that is aimed at creating a comprehensive model to study laryngeal reinnervation following injury.
A number of contemporary groups have made attempts at studying neuroregenerative agents in rats
after injury.11-13 Although some of this work holds
potential promise, most of these studies are based on
incomplete animal models (without functional studies and sometimes without experimental controls),
leaving room for doubt about their validity.
Understanding reinnervation requires examination of events at the site of injury (the nerve) and
the effects on the end organ (the larynx), and also
an understanding of events at the level of the motor
neurons (the NA). For laryngeal nerve injury, this
is a complicated proposition, given contributions to
the larynx from both the SLN and the RLN, with
the RLN carrying both adductory and abductory efferent fibers. Cataloging the organization of LMNs
within the NA is of paramount importance in understanding the potential reorganization of abductor
and adductor signals that can occur after injury leading to synkinesis. Once the adductor and abductor
LMN populations are established in normal animals,
one could look for inappropriate LMN labeling in
the LMNs in the NA. Conversely, the possibility exists that an axon of one LMN may innervate both
muscles. In these scenarios, one would see double
labeling of LMN with two different dyes. For these
reasons, it is critical to establish the accurate position of the LMN pool for the RLN and the SLN.
Our future studies stand to unravel the varied populations of LMNs that innervate separate sets of muscles in the larynx.
The study of the RLN and SLN LMNs in rats is
not novel. Using horseradish peroxidase (HRP), diamidino yellow, and true blue, Portillo and Pásaro1
injected laryngeal muscle groups. For the cricothy-
roid muscle, 22 to 90 LMNs were labeled, depending on the labeling agent used. The mean value for
the posterior cricoarytenoid, thyroarytenoid, and
lateral cricoarytenoid muscles combined was 215.1
Therefore, working under the assumptions that the
cricothyroid LMNs constitute the LMNs of the SLN
and that the sum of the posterior cricoarytenoid, thyroarytenoid, and lateral cricoarytenoid LMNs equal
those of the RLN, the mean values of labeled LMNs
in their study were lower than those in the present
study. In another study, Hinrichsen and Ryan2 used
HRP to label the cut nerve ends of the RLN, and
they immersed the nerve endings in an HRP solution, thereby increasing cell labeling; however, significant variability in results was encountered. Both
studies1,2 used labeling agents that are somewhat
unreliable and are likely the source of much of the
difference and confusion in the literature. Furthermore, injection of HRP into muscles as opposed to
labeling transected nerve ends has the potential to
label fewer LMNs.
Flint et al3 undertook a similar approach to categorize the LMNs and addressed questions regarding
postinjury synkinesis in animals. Horseradish peroxidase, diamidino yellow, and fast blue were used
to label either specific muscle groups or nerves. The
relationships of the LMN muscle groups were largely consistent with those in previous studies; however, labeling of the RLN showed fewer LMNs than in
similar studies and the present study. In addition, an
experimental condition was undertaken in which the
posterior cricoarytenoid, thyroarytenoid, and lateral
cricoarytenoid muscles were injected with HRP 15
weeks after transection and reanastomosis. These authors showed a decreased number of labeled LMNs,
as well as a loss of spatial segregation in respect to
the studied LMN groups, validating the importance
of analyzing the LMNs when considering laryngeal
nerve injury and synkinesis.
The current study used FG and FR, two examples
of retrograde fluorolabeling agents. Previous work
in rats using biotinylated dextran amines (BDAs) as
labeling agents has generated results similar to those
of the current study. When BDAs were used to label
the RLN, there was a range of 121 to 240 LMNs,
with an average of 143.4 Again using BDAs, Pascual-Font et al5 found that the SLN had an average
of 111 LMNs. In addition to SLN labeling, they noted significant labeling of the ipsilateral dorsal motor nucleus of the vagus nerve — a finding attributed to efferent preganglionic parasympathetic neurons. Additional labeling was noted in the ipsilateral
solitary tract and the nucleus of the ipsilateral solitary tract, both likely representing afferent laryngeal
sensory fibers carried within the SLN.5 The present
Weissbrod et al, Recurrent & Superior Laryngeal Nerve Lower Motor Neurons
study confirms the above LMN counts, although our
labeling was more consistent. We also noted very
few labeled cells outside the expected confines of
the NA; however, we did not quantify these cells, as
they do not represent LMNs that drive the RLN or
SLN and do not contribute to postinjury synkinesis.
In the present series of experiments, we found
only that FR reliably labels the SLN without contamination. With FR, the mean number of labeled
neurons was 116, with a range of 74 to 158. These
results are consistent with those of prior studies.1,4,5
The disparity within fluorolabeling agents is consistent with a number of previous studies that have
shown that FR only effectively labels damaged or
cut nerves in the facial muscles of the rat,14 and that
it has poor labeling capacity when injected directly
into muscle.15 This tendency would make FR contamination via surrounding musculature unlikely.
Conversely, in a mouse model of the tibial nerve
and gastrocnemius muscles, FG showed good retrograde transport in both cut nerve labeling and muscle injection, but was found to show an increased
potential for background diffusion. These findings
suggest that if muscle is damaged during surgery,
contamination and resultant LMN labeling with FG
is more likely.
In the present study, LMN labeling outside of the
NA can be attributed to a number of possible scenarios. First, there are afferent sensory and parasympathetic nerves within the RLN and SLN that originate in the nucleus of the solitary tract and the dorsal
motor nucleus. It is further possible that contamination of surrounding tissues takes place during the
dissection of the RLN and SLN. Access to the SLN
requires significantly more dissection of surrounding muscles than does access to the RLN. There are
85
fascial planes over all of the surrounding structures
that can serve as natural barriers to contamination.
As such, contamination with FG via injured muscle
is likely the reason for the disparate SLN quantification in our study that resulted in elevated numbers of
LMNs. In the present study, regardless of the agent
used, the RLN LMNs were consistently labeled in
number and location.
In conclusion, labeling of the RLN and SLN, with
FR in particular, produces consistent results that allow the quantification and confirmation of the position of the LMNs of the laryngeal nerves. FluoroRuby is optimal for this study because of its ardent
ability to be taken up by transected nerves with minimal contamination of surrounding tissues. Although
FG is also avidly taken up by cut nerves, we believe
it also has the tendency to contaminate nearby tissues in situations in which there has been extensive
muscle dissection. The position of the LMNs of the
RLN and SLN in this study is consistent with the
findings of prior studies: the LMNs of the SLN lie
rostral, ventral, and lateral to those of the RLN, with
a small area of overlap. This study also confirms the
higher number of RLN LMNs that were identified
in one study4 and the location of these LMNs identified in others.3,6 The present observations support
the superiority of the new staining techniques and
resolve the controversy and confusion that resulted
from disparate LMN counts seen in studies that used
other techniques. The information from this study
further clarifies the use of the rat as a model for
RLN injury. This study also provides baseline information for future study of specific intralaryngeal
muscle LMN groups and, eventually, the study of
neurotropic or other pharmacologic agents with the
motive of improving postinjury function and reducing synkinesis.
Acknowledgment: We thank Dr Alan Springer for his statistical analysis.
REFERENCES
1. Portillo F, Pásaro R. Location of motoneurons supplying the intrinsic laryngeal muscles of rats. Horseradish peroxidase and fluorescence double-labeling study. Brain Behav Evol
1998;32:220-5.
5. Pascual-Font A, Maranillo E, Merchán A, Vázquez T,
Sañudo JR, Valderrama-Canales FJ. Central projections of the
rat superior laryngeal nerve [in Spanish]. Acta Otorrinolaringol
Esp 2006;57:295-9.
2. Hinrichsen CFL, Ryan AT. Localization of laryngeal motoneurons in the rat: morphologic evidence for dual innervation? Exp Neurol 1981;74:341-55.
6. Bieger D, Hopkins DA. Viscerotopic representation of
the upper alimentary tract in the medulla oblongata in the rat:
the nucleus ambiguus. J Comp Neurol 1987;262:546-62.
3. Flint PW, Downs DH, Coltrera MD. Laryngeal synkinesis following reinnervation in the rat. Neuroanatomic and physiologic study using retrograde fluorescent tracers and electromyography. Ann Otol Rhinol Laryngol 1991;100:797-806.
7. Ahmed FAKM, Ingoglia NA, Sharma SC. Axon resealing following transection takes longer in central axons than in
peripheral axons: implications for axonal regeneration. Exp
Neurol 2001;167:451-5.
4. Pascual-Font A, Maranillo E, Vázquez T, Sañudo JR,
Valderrama-Canales FJ. On the number and morphometrical
parameters of the nucleus ambiguous neurons after the injury
and regeneration of the recurrent laryngeal nerve in the rat [in
Spanish]. Acta Otorrinolaringol Esp 2008;59:163-9.
8. Tessema B, Roark RM, Pitman MJ, Weissbrod P, Sharma
S, Schaefer SD. Observations of recurrent laryngeal nerve injury
and recovery using a rat model. Laryngoscope 2009;119:164451.
9. Tessema B, Pitman MJ, Roark RM, Berzofsky C, Sharma
86
Weissbrod et al, Recurrent & Superior Laryngeal Nerve Lower Motor Neurons
S, Schaefer SD. Evaluation of functional recovery of recurrent
laryngeal nerve using transoral laryngeal bipolar electromyography: a rat model. Ann Otol Rhinol Laryngol 2008;117:604-8.
10. Pitman MJ, Weissbrod P, Roark R, Sharma S, Schaefer
SD. Electromyographic and histologic evolution of the recurrent laryngeal nerve from transection and anastomosis to mature
reinnervation. Laryngoscope 2011;121:325-31.
11. Shiotani A, O’Malley BW Jr, Coleman ME, Flint PW.
Human insulinlike growth factor 1 gene transfer into paralyzed
rat larynx: single vs multiple injection. Arch Otolaryngol Head
Neck Surg 1999;125:555-60.
12. Mori Y, Shiotani A, Saito K, et al. A novel drug therapy
for recurrent laryngeal nerve injury using T-588. Laryngoscope
2007;117:1313-8.
13. Hydman J, Remahl S, Björck G, Svensson M, Mattsson
P. Nimodipine improves reinnervation and neuromuscular function after injury to the recurrent laryngeal nerve in the rat. Ann
Otol Rhinol Laryngol 2007;116:623-30.
14. Choi D, Li D, Raisman G. Fluorescent retrograde neuronal tracers that label the rat facial nucleus: a comparison of
Fast Blue, Fluoro-ruby, Fluoro-emerald, Fluoro-Gold, and DiI.
J Neurosci Methods 2002;117:167-72.
15. Hayashi A, Moradzadeh A, Hunter DA, et al. Retrograde
labeling in peripheral nerve research: it is not all black and
white. J Reconstr Microsurg 2007;23:381-9.
Assessment of Canine Vocal Fold Function After Injection of a
New Biomaterial Designed to Treat Phonatory Mucosal Scarring
Sandeep S. Karajanagi, PhD; Gerardo Lopez-Guerra, MD; Hyoungshin Park, PhD;
James B. Kobler, PhD; Marilyn Galindo; Jon Aanestad; Daryush D. Mehta, PhD;
Yoshihiko Kumai, MD, PhD; Nicholas Giordano; Anthony d’Almeida;
James T. Heaton, PhD; Robert Langer, ScD; Victoria L. M. Herrera, MD;
William Faquin, MD, PhD; Robert E. Hillman, PhD; Steven M. Zeitels, MD
Objectives: Most cases of irresolvable hoarseness are due to deficiencies in the pliability and volume of the superficial
lamina propria of the phonatory mucosa. By using a US Food and Drug Administration–approved polymer, polyethylene glycol (PEG), we created a novel hydrogel (PEG30) and investigated its effects on multiple vocal fold structural and
functional parameters.
Methods: We injected PEG30 unilaterally into 16 normal canine vocal folds with survival times of 1 to 4 months. Highspeed videos of vocal fold vibration, induced by intratracheal airflow, and phonation threshold pressures were recorded
at 4 time points per subject. Three-dimensional reconstruction analysis of 11.7 T magnetic resonance images and histologic analysis identified 3 cases wherein PEG30 injections were the most superficial, so as to maximally impact vibratory
function. These cases were subjected to in-depth analyses.
Results: High-speed video analysis of the 3 selected cases showed minimal to no reduction in the maximum vibratory
amplitudes of vocal folds injected with PEG30 compared to the non-injected, contralateral vocal fold. All PEG30-injected vocal folds displayed mucosal wave activity with low average phonation threshold pressures. No significant inflammation was observed on microlaryngoscopic examination. Magnetic resonance imaging and histologic analyses revealed
time-dependent resorption of the PEG30 hydrogel by phagocytosis with minimal tissue reaction or fibrosis.
Conclusions: The PEG30 hydrogel is a promising biocompatible candidate biomaterial to restore form and function to
deficient phonatory mucosa, while not mechanically impeding residual endogenous superficial lamina propria.
Key Words: hoarseness, hydrogel, polyethylene glycol, scar.
INTRODUCTION
there has not been a reliable method to restore normal suppleness to stiff vocal folds.7,8 This capability
would be highly desirable, given the large numbers
of patients with benign and malignant processes
who sustain diminished pliability of phonatory mucosa from phonotrauma, disease, or instrumentation.
About 6.6% of the American working-age population is affected by voice-related disorders, and most
of these patients present with diminished phonatory
Optimal vocal function requires an aerodynamically competent glottal valve and pliable phonatory
mucosa. During the past century, surgeons became
adept at repairing aerodynamic incompetence by restoring glottal closure. This was achieved by augmenting the paraglottic space1-4 and/or repositioning
the arytenoid5,6 to close the rima glottidis. However,
From the Center for Laryngeal Surgery and Voice Rehabilitation (Karajanagi, Lopez-Guerra, Park, Kobler, Galindo, Aanestad, Mehta,
Kumai, d’Almeida, Heaton, Hillman, Zeitels) and the Department of Pathology (Faquin), Massachusetts General Hospital, the Departments of Surgery (Karajanagi, Lopez-Guerra, Park, Kobler, Kumai, Heaton, Hillman, Zeitels) and Pathology (Faquin), Harvard Medical School, the Department of Medicine, Boston University School of Medicine (Herrera), the School of Engineering and Applied
Sciences, Harvard University (Mehta), and the School of Engineering, Boston University (Giordano), Boston, and the Department of
Chemical Engineering (Karajanagi, Park, Langer) and the Harvard-MIT Division of Health Sciences and Technology (Kobler, Heaton,
Langer, Hillman), Massachusetts Institute of Technology, Cambridge, Massachusetts. This work was supported in part by the Eugene
B. Casey Foundation and the Institute of Laryngology and Voice Restoration. Dr Zeitels has an equity interest in Endocraft LLC. This
study was performed in accordance with the PHS Policy on Humane Care and Use of Laboratory Animals, the NIH Guide for the Care
and Use of Laboratory Animals, and the Animal Welfare Act (7 U.S.C. et seq.); the animal use protocol was approved by the Institutional Animal Care and Use Committee (IACUC) of the Massachusetts General Hospital.
Presented at the meeting of the American Broncho-Esophagological Association, Las Vegas, Nevada, April 28-29, 2010. Winner of the
Broyles-Maloney Award.
Correspondence: Sandeep S. Karajanagi, PhD, Center for Laryngeal Surgery and Voice Rehabilitation, Massachusetts General Hospital, Bartlett Hall Extension, 413, Boston, MA 02114.
87
88
Karajanagi et al, Canine Vocal Fold Function After Injection of New Biomaterial
mucosal pliability of their vocal folds.9
The need for methods to restore vocal fold pliability has generated an increasing number of efforts
to develop biological and/or synthetic remedies. A
recent series of review articles provides an excellent
overview of the wide range of treatment strategies
that are under development.10-17 These proposed
therapies include the use of synthetic biomaterials
(some based on natural extracellular matrix components such as hyaluronic acid [HA] and collagen),
cells (stem cells or fibroblasts), autologous tissue
(fat or fascia), scaffolds (synthetic or tissue-based),
and growth factors and other chemical agents, such
as steroids and phytochemicals. All of these approaches have demonstrated some positive results
in animal studies, and small-scale human trials have
studied several types of treatment.
Biomaterials have several potential advantages
over other approaches, including availability, limited biological side-effects, control over viscoelastic material properties, immediate benefit, and procedural simplicity (eg, delivery via injection). Recently, some human subject experiments suggested
that biomaterial therapy may be effective for vocal
fold scar. In a 2-year study, 40 patients with vocal
fold scar, vocal fold atrophy, or sulcus vocalis were
injected with auto–cross-linked polysaccharide HA
gel (Fidia Advanced Biopolymers, Abano Terme,
Padova, Italy).18 The material was placed into the
lamina propria for treating the scar and sulcus or
into the thyroarytenoid muscle for closing the glottal gap. This study had a pretreatment-posttreatment
design, with multivariate statistical analysis of voice
and endoscopic parameters. Glottal closure, vibratory pattern, and mucosal waves were significantly improved at the first follow-up (1 to 3 months)
and at 12 months after injection. In two other studies
using esterified HA (MeroGel; Medtronic Xomed,
Jacksonville, Florida), the placement of HA-based
fibers beneath phonosurgical microflaps appeared to
have a positive effect on the long-term measures of
voice production.19,20
The positive vocal outcomes reported in these initial human experiments are somewhat tempered by
the heterogeneity of the vocal conditions treated and
by inadequate controls. However, this work, as well
as a host of animal studies,13,21-23 does suggest that
biomaterials may provide short- and long-term benefits for treatment of vocal fold scar, and that materials with established biocompatibility in other parts
of the body can be relatively safe for use in the vocal folds.
Hylan-B, auto-crosslinked polysaccharide HA
gel, and MeroGel were initially designed for appli-
cation in other tissues (eg, skin or dermis), and it
would be serendipitous if they happened to be optimal for vocal fold applications. Ideally, substances
would be designed specifically for the vocal fold on
the basis of the rheological and functional properties
of the normal vocal fold mucosa, as well as for their
use in scar-stiffened tissue. Since the primary component of the vocal fold’s layered microstructure responsible for vibration is the superficial lamina propria (SLP),24 we created a novel hydrogel, PEG30
(patent pending), with rheological properties that
lie near the lower limits of normal vocal fold stiffness. In choosing materials, an additional consideration becomes the potential regulatory hurdles for
approval of novel chemical formulations. We therefore based PEG30 on polyethylene glycol (PEG) because PEG is an established biocompatible polymer
widely used in numerous products approved by the
US Food and Drug Administration. We investigated
its effects on multiple vocal fold structural and functional parameters.
MATERIALS AND METHODS
Preparation of PEG30 Hydrogel. PEG30 hydrogels were prepared by photopolymerization of PEG
diacrylate (PEG-DA; SunBio Inc, Orinda, California) in the presence of PEG (Aldrich, St Louis, Missouri; both with molecular weight of 10 kd). Briefly,
aqueous solutions of PEG and PEG-DA (both 100
mg/mL) were made in sterile phosphate-buffered
saline solution (PBS) and mixed in the volumetric
ratio 3:7 of PEG-DA to PEG. This solution (1 mL)
was gelled for 200 seconds with 0.05% (wt/vol) of
Irgacure 2959 (Ciba, Tarrytown, New York) as the
photoinitiator. An EXFO Lite lamp (Lumen Dynamics Group, Mississauga, Canada) that generated ultraviolet light of intensity 10 mW/cm2 (measured at
365 nm) was used for the gelation. The entire process was carried out in a sterile laminar flow hood.
The gel disks thus formed were rinsed with 70%
ethanol (3× at 1 minute per wash), rinsed in sterile
PBS (3× at 5 minutes per wash), and then incubated
in sterile PBS for 24 hours at 37°C. The swollen gels
were progressively sheared through needles of decreasing bore size to make them injectable through a
25-gauge needle. The gels were stored at 4°C sealed
in a capped syringe until further use.
Mechanical Testing of Hydrogels. The viscoelastic shear properties of the hydrogels were measured
at low frequencies with an AR-2000 rheometer (TA
Instruments, Inc, New Castle, Delaware). A coneand-plate geometry was used to apply oscillatory
shear to hydrogel samples with an acrylic cone (60mm diameter, 2° angle) and a flat metallic Peltier
plate heated to 37°C. Sheared gels were placed be-
Karajanagi et al, Canine Vocal Fold Function After Injection of New Biomaterial
tween the heated plate and the cone so that a manufacturer-specified gap of 61 μm was maintained
between the cone and the plate. The gel was subjected to an oscillatory shear at 1 Hz for 2 minutes
to equilibrate the entire gel to a uniform temperature of 37°C. Strain sweep tests were done to ensure
that the shear property measurements were done in
the linear region of the stress-strain curve. The viscoelastic shear properties are independent of the
percentage strain in the linear region. A target shear
strain value was therefore identified by measuring
the viscoelastic shear properties as a function of the
percentage strain applied. A 0.6% strain was typically used to measure the shear properties by means
of a frequency sweep. Measurements of the shear
properties were then made by systematically varying the frequency from 1 to 10 Hz. The elastic shear
modulus (G') at 10 Hz was used as a measure of the
mechanical properties of the PEG30 hydrogel used
in this study. The elastic shear modulus of PEG30
fell within or below the ranges reported in the literature for normal phonatory mucosa,25 thereby demonstrating adequate softness and/or pliability at low
frequencies to justify in vivo testing.
Animal Care. This study was performed in accordance with the PHS Policy on Human Care and Use
of Laboratory Animals, the NIH Guide for the Care
and Use of Laboratory Animals, and the Animal
Welfare Act (7 U.S.C. et seq.). All experimentation
followed an animal use protocol that was approved
by the Committee on Animal Care and the Institutional Animal Care and Use Committee of the Massachusetts General Hospital. All surgical procedures
and housing of animals took place at the Massachusetts General Hospital.
Sixteen research-bred dogs (young adult male
hounds, 8 months to 1 year in age) were obtained
from Covance Research Products (Cumberland, Virginia). Through an arrangement with the supplier, we
were able to obtain animals that had been endoscopically pre-screened for laryngeal disorders, which
are not uncommon due to excessive barking. Before
every procedure, the dogs were premedicated with
acepromazine maleate (0.05 mg/kg), glycopyrrolate (0.01 mg/kg), and butorphanol tartrate (0.2 mg/
kg). Anesthesia was maintained throughout the surgical procedures with intravenous propofol (10 mL
for induction, followed by 0.5 mg/kg per minute or
more given intravenously as needed). Spontaneous
respiration was maintained, medical-grade oxygen
was supplied continuously through the mouth, and
a heating pad was used to maintain body temperature throughout the surgery. Heart and respiratory
rates were monitored continuously by a monitoring
89
device and visual observation. At the conclusion of
each phonatory testing session (see below), the dogs
were watched until they regained consciousness and
were given the nonsteroidal anti-inflammatory drug
meloxicam (0.2 mg/kg), buprenorphine hydrochloride (0.01 mg/kg), and the antibiotic Baytril (enrofloxacin; 5 mg/kg).
Surgical Procedures. The dogs were placed in a
supine posture on a surgical table with the head and
neck supported. The vocal folds were exposed by
standard endoscopic techniques and visualized with
a Leica F40 surgical microscope. PEG30 was injected unilaterally in 16 normal canine vocal folds
with postinjection survival periods of 1, 2, 3, and 4
months (n = 4 per time point). The contralateral fold
served as a control. An average of 60 μL of PEG30
was injected in the vocal fold with a 25-gauge Zeitels Vocal-Fold Infusion Needle26 (Endocraft, Providence, Rhode Island) and a specially modified industrial viscous fluid dispenser (model HP7× by
Nordson EFD, Westlake, Ohio) that was pneumatically driven and provided improved injection control compared with conventional handheld syringes
or injection guns. The HP7× handpiece was modified to use a high-precision linear transducer and
digital meter so that dispenser output could be measured in real time. The injection volumes for dogs 1
to 3 reported in this study were 69, 52, and 53 μL,
respectively.
We made periodic examinations of in vivo vocal
fold appearance using standard microlaryngoscopy.
Specifically, examinations were made at the following time points after injection for dogs with 1-, 2-, 3-,
and 4-month survival times, respectively: 1, 2, and
4 weeks; 2, 5, and 8 weeks; 4, 8, and 12 weeks; and
4, 8, 12, and 16 weeks. High-speed videos (4,000
frames per second) of the vocal fold vibration were
also recorded during these examinations by using
a 16-gauge needle inserted in the trachea to create
subglottic air pressure for phonation. Air delivered
for phonation was warmed (37°C to 39°C) and humidified by a Conchatherm unit (high-flow model;
Hudson RCI, Temecula, California). A pressure sensor (MPX2010GP; Motorola, Schaumburg, Illinois)
was mounted on the air supply tube near its connection to the needle inserted into the trachea, and
a condenser microphone (ECM50PSW; Sony, New
York, New York) was positioned approximately 15
cm from the mouth opening. Simultaneous acoustic
and pressure signals were filtered and recorded digitally (20,000-Hz sampling rate) with Axon Instruments hardware (Cyberamp 380, Digidata 1440a)
and software. Phonation threshold pressures were
monitored in real time during data collection in or-
90
A
Karajanagi et al, Canine Vocal Fold Function After Injection of New Biomaterial
B
Fig 1. Example endoscopic view of vocal folds and derivation of amplitude asymmetry measure. A) High-speed video image
taken during maximum abduction during phonation. B) Derivation of lateral displacement waveforms for right and left vocal
folds. Images are rotated to orient glottis vertically and are cropped to optimize image processing. Digital kymogram is generated from position halfway between anterior and posterior commissures. Lateral displacement waveforms are then obtained by
tracing edges of vocal folds in digital kymogram. Relative vocal fold pliability is estimated by calculating ratio between maximum amplitude of vocal fold injected with biomaterial and that of noninjected vocal fold.
der to monitor and control phonation driving pressures, and were also measured from recorded signals
during subsequent analyses. Our method for testing
vocal function uses a tracheal needle to supply air
with instrument-manipulated adduction of the vocal
folds to initiate phonation. These recordings allow
us to assess the pliability of the vocal folds under
physiological conditions. Furthermore, this in vivo
procedure allows us to test vocal function at multiple time points, which is ideal for following the
progress of the effect of PEG30 injection over time
and minimizes the number of animals needed.
High-Speed Video Data Collection and Analysis. Imaging of canine vocal fold vibration during
induced phonation was accomplished by attaching a Phantom v7.3 high-speed video camera (Vision Research, Inc, Wayne, New Jersey) to the optical adapter of the operating microscope. This enabled magnified high-quality color imaging at 12bit quantization with a complementary metal–oxide–semiconductor image sensor. High-speed video
data were recorded at 4,000 images per second with
maximum integration time and a spatial resolution
of 448 horizontal × 424 vertical pixels to capture an
approximately 2-cm2 target area. Illumination was
provided by a Leica F40 surgical microscope with
an integrated 300-W xenon arc light source. Each
high-speed video data segment consisted of 1,000
images (250 ms).
A previous report describes the digital image processing methods that were used to estimate lateral
displacements of the left and right vocal folds from
the recorded high-speed video segments.27 Briefly,
the processing steps included correction of motion
artifact; user selection of a glottal midline; rotation
and cropping of images to vertically orient the midline; conversion from color to monochromatic images; user selection of an intensity threshold that optimized glottal edge detection; generation of a digital
kymogram at the mid-musculomembranous glottis;
and extraction of waveforms representing the amplitude of left and right vocal fold motion from the
kymograms (Fig 1). To assess the impact of the injected biomaterial on the pliability of the vocal fold,
we defined a measure of left-right amplitude asymmetry as the ratio between the maximum amplitude
of the injected vocal fold and the maximum amplitude of the noninjected vocal fold.
Magnetic Resonance Microscopy and/or Microimaging. Immediately after euthanasia, the larynges were removed, rinsed with 0.9% saline solution,
and fixed in cold PBS-buffered 4% paraformaldehyde. Magnetic resonance microimaging of the excised larynges was done as described elsewhere.28
Briefly, the larynges were suspended in Fomblin
and imaged with a Bruker Avance-500 nuclear magnetic resonance spectrometer (11.7 T; 500 MHz for
proton). A 30-mm “birdcage” transmitter and/or receiver coil was used for imaging. Transverse and/or
coronal scans were obtained throughout the length
of the segment with multi-slice multi-echo sequences. Transverse and/or coronal and axial images were
then obtained through regions of interest. Gradient
recalled echo images were acquired according to
the following parameters: slice thickness, 0.3 mm;
field of view, 20 mm; MTX, 512 (pixel size, 39 μm);
and the following pulse parameters: repetition time,
1,000 ms; echo time (effective), 8.3 ms; flip angle,
10°; and NEX, 32. Sixty-four transverse or coronal slices and 32 axial slices were obtained per dog
larynx for all 16 dogs injected. Fat was suppressed
Karajanagi et al, Canine Vocal Fold Function After Injection of New Biomaterial
91
Fig 2. Representative example of histology–
magnetic resonance imaging matching (top
panels) that was used for 3-dimensional reconstruction of vocal fold with Amira software (bottom panels). Arrows show location
of residual PEG30 and cellular infiltrate. A —
anterior; P — posterior.
with a fat-presaturation pulse during imaging. Data
were processed with Paravision software provided
by the vendor.
Histologic Analysis. Each excised larynx was bisected in the sagittal plane, and each half was then
trimmed and cut in two pieces in the coronal plane
near the middle of the vocal folds. The pieces were
oriented appropriately and embedded in paraffin.
Coronal sections, 5 μm thick, were cut and saved
at fixed intervals along the anterior-posterior extent of the vocal fold. For each specimen, we used
the magnetic resonance images to guide the cutting of the paraffin sections such that the injection
sites were fully encompassed. Alternate slides were
stained with hematoxylin and eosin and Masson’s
trichrome stains and examined with brightfield microscopy. Digital photomicrographs were obtained
with a Zeiss Axioskop2 microscope system. When
necessary, differential interference contrast microscopy was used to improve image quality.
Identification of Cases With Superficial Injections. In order to correctly attribute potential postin-
jection changes in the in vivo vibration of the vocal folds to the injected biomaterial, we reasoned
that it would be important that the injected biomaterial and the reaction that it caused be extremely
superficial so as to maximally affect vocal fold vibration. In order to visualize and quantify the 3-dimensional (3-D) location of the residual biomaterial
and the accompanying biological reaction, we used
calibrated magnetic resonance images in combination with histologic imaging. Masson’s trichrome–
stained histologic sections were matched with coronal magnetic resonance microimaging slices in
order to delineate residual biomaterial and cellular
infiltrate from the surrounding vocal fold tissue in
the magnetic resonance microimages as described
in detail elsewhere.28 An example of magnetic resonance imaging–histology image matching is shown
in Fig 2 (top panels). Three-dimensional reconstruction of consecutive calibrated magnetic resonance
microimaging slices was then done with Amira software (Visage Imaging, Inc, San Diego, California)
to visualize the location and quantify the volume of
the residual biomaterial and cellular infiltrate (Fig 2,
92
Karajanagi et al, Canine Vocal Fold Function After Injection of New Biomaterial
Fig 3. “Vibratory zone” (shaded in blue) of vocal fold
identified in magnetic resonance microimaging slice of
vocal fold. Arrow shows location of residual material and
cellular infiltrate.
bottom panels).
Data from all 16 cases were screened to identify
subjects that had residual biomaterial and/or cellular infiltrate in the key “vibratory zone” of the vocal
folds. The vibratory zone of the vocal fold was identified as the area in the SLP defined by points 1 mm
on the superior surface and 2.5 mm on the medial
surface from the free edge of the vocal fold (Fig 3).
As part of the screening process, subjects that had
less than 5 μL of residual biomaterial and cellular
infiltrate were eliminated. For each of the remaining
subjects, 3 to 4 consecutive magnetic resonance microimaging slices with the highest percentage of residual biomaterial and cellular infiltrate were identified. For each of these magnetic resonance microimaging slices, we computed the “vibratory fraction,”
ie, the percentage of the vibratory zone occupied by
the residual biomaterial and cellular infiltrate, and
averaged it across the measured slices. The 3 animals that had the highest average vibratory fractions
were identified and subjected to in-depth analyses.
Representative magnetic resonance microimages
and the 3-D reconstructions of vocal folds for the 3
selected cases are shown in Fig 4. One subject each
with survival time points of 1 month, 2 months, and
3 months constituted the 3 subjects selected by this
screening process.
RESULTS
Vocal Fold Function Before and After PEG30
Injection. We evaluated vocal fold function for the
3 selected animals (those with the most superfi-
cial PEG30 injections; see Methods) during in vivo
phonation, whereby delivery of subglottic air pressure and manual adduction of the arytenoid cartilages caused the vocal folds to valve the airstream
in a manner resembling natural phonation. All vocal folds at all time points displayed clear evidence
of mucosal wave activity. The average phonation
threshold pressures were low (4 to 8.5 cm H2O).
Analysis of symmetry in vocal fold amplitude failed
to show a consistent difference in lateral tissue excursion for the injected versus noninjected (control)
vocal folds (Fig 5). Statistical comparison of the relative vocal fold amplitude (injected divided by noninjected) prior to injection versus the last recording time for each animal failed to find a difference
(2-tailed paired t-test, p = 0.26). Likewise, phonation threshold pressures did not differ between preinjection measures and the last recording session for
each animal (2-tailed t-test, p = 0.40; Fig 6). Taken
together, these two findings suggest that the injected
gel and/or tissue response did not appreciably alter
the pliability of the superficialmost portion of the
SLP during manually elicited phonation.
Biocompatibility of PEG30. Multiple in vivo examinations of the canine vocal folds with an operating microscope and gentle blunt-instrument palpation revealed no clinical evidence of inflammation
such as erythema or edema. In all 3 cases, the injected vocal fold surface looked identical to the noninjected vocal fold under high magnification. Histologic analysis of the vocal fold for canine 1 (survival
time of 1 month after injection) revealed the presence of PEG30 in the vocal fold, accompanied by
significant macrophage infiltration at the implantation site, including and surrounding the PEG30 (Fig
7A,B). To a lesser extent, lymphocytes and plasma
cells were also present. No giant cells or neutrophils
were observed. Slight to mild fibrosis was seen, but
no fibrotic capsule formation was observed around
the PEG30. The macrophages adjacent to the implant
had a foamy cytoplasm suggesting active phagocytosis of the PEG30 by the macrophages. These observations suggest that PEG30 is biodegradable by
macrophage-mediated phagocytosis. Similar histopathologic findings were observed for canine 2 (survival time of 2 months after injection) and canine 3
(survival time of 3 months after injection), although
with amounts of residual PEG30 and macrophages that decreased with each successive month (Fig
7C,D). No fibrosis in the vocal fold was seen for either canine 2 or canine 3. The foreign body response
to PEG30 seemed to resolve over time, with no apparent damage to the surrounding vocal fold tissue
and with replacement of the lost material by loose
connective tissue.
Karajanagi et al, Canine Vocal Fold Function After Injection of New Biomaterial
93
Fig 4. Representative magnetic resonance microimages (top panels) and respective 3-dimensional reconstructions (bottom panels) of vocal folds for 3 selected dogs. “Vibratory zone” is shaded in blue, and arrows show location of residual PEG30 and cellular infiltrate. Canine 1 received 2 injections, both at time zero. Relatively more superficial-medial injection is shown by arrow
in magnetic resonance microimage and in 3-dimensional reconstruction. A — anterior; P — posterior.
In all 3 animals, no signs of any long-lasting fibrosis or other tissue damage were seen. Overall, the
absence of a neutrophil response, the lack of a fibrotic capsule, and the macrophage-mediated clearance of PEG30 with minimal to no fibrosis suggest
that PEG30 is biocompatible and biodegradable in
Fig 5. Comparison of ratios of amplitude asymmetry between injected and noninjected vocal folds for 3 selected
dogs as function of time.
the vocal folds.
DISCUSSION
Normal vocal fold vibration is manifested primarily as a wave of displaced mucosal tissue on the sur-
Fig 6. Comparison of phonation threshold pressures for 3
selected dogs as function of time.
94
Karajanagi et al, Canine Vocal Fold Function After Injection of New Biomaterial
A
B
C
D
face of the vocal folds, ie, the mucosal wave. The
phonatory or vibratory mucosa is mostly composed
of a soft and pliable layer of SLP with a thin covering of epithelium that essentially mimics the biomechanical properties of the SLP substrate. The main
cause of chronic dysphonia or voice loss is permanent damage and/or scarring of the normal SLP. No
proven methods currently exist for restoring normal
function to vocal folds that have lost the pliability of
this key vibratory layer.
This report describes the results from initial testing of a new synthetic PEG30 hydrogel being developed to restore vibratory function to vocal folds that
have lost some pliability of the phonatory mucosa.
Polyethylene glycol was chosen as the basis for this
new material because numerous PEG-based materials are already approved by the US Food and Drug
Administration for implants in other parts of the
body, and therefore our modification had good potential to be biocompatible. In addition, during initial low-frequency rheometric screening, the PEG30
formulation seemed soft and pliable enough to potentially mimic the biomechanical function of normal SLP, and it could be injected through the different types of 25-gauge needles currently used for
vocal fold infusion during phonosurgery.26 It was
Fig 7. Masson’s trichrome–
stained images of PEG30-injected vocal folds of A,B) canine 1,
C) canine 2, and D) canine 3.
deemed necessary to first determine if the impact
of PEG30 on healthy vocal fold tissue and function
was acceptable prior to investigating its use in vocal
fold pathology, particularly since most patients who
have lost some pliability of the phonatory mucosa
still retain some normal regions of SLP.
The biocompatibility and impact on vocal function of PEG30 were tested longitudinally in an in
vivo canine model with survival times ranging from
1 to 4 months. This report focuses on results from 3
dogs (1 each at 1, 2, and 3 months) for whom 3-dimensional reconstruction analysis of 11.7-T magnetic resonance imaging and serial-section histologic analysis of excised larynges showed that the
PEG30 injections were superficial, so as to ensure
maximal impact on vocal fold vibratory function.
The presence of PEG30 did not negatively affect
normal phonatory vibration: the injected canine
vocal folds displayed clear mucosal waves at all 3
postinjection time points, vibratory amplitudes that
did not differ significantly from those of the contralateral noninjected vocal folds, and no significant
change in phonation threshold pressure relative to
preinjection phonation.
The PEG30 also appeared to elicit a minimal biological response when implanted in the SLP of ca-
Karajanagi et al, Canine Vocal Fold Function After Injection of New Biomaterial
nine vocal folds, with no evidence of an overt inflammatory response either in terms of the vocal
fold surface or with respect to histologic findings.
The biological response to PEG30 in the vocal folds
of the remaining 13 dogs was similar to that seen in
the 3 selected cases. In all dogs, the residual PEG30
was actively phagocytosed by macrophages, and the
amount of residual PEG30 and macrophages progressively decreased with an increase in survival
period. The PEG30 and the foreign body reaction
were resorbed with no apparent damage to the surrounding vocal fold tissue, and the lost PEG30 was
replaced by nonfibrotic loose connective tissue. An
in-depth examination of the biological response
to PEG30 in all 16 animals, however, is out of the
scope of this report that focuses primarily on the
functional evaluation of PEG30 injected in the phonatory mucosa. A future report will utilize histologic
data from the entire cohort of 16 animals to evaluate
the biological response to PEG30 in detail.
Overall, comparative analysis of histologic results across 1- to 3-month survival end points indicates that after PEG30 is injected into the SLP of vocal folds it biodegrades within 3 to 4 months without creating damage (fibrosis) or mechanical changes (stiffness) in healthy tissue. If it is assumed that
there are no residual beneficial effects after PEG30
has been resorbed (eg, stimulation of tissue remod-
95
eling), then the current formulation of PEG30 would
probably have to be reinjected at approximately
3-month intervals to maintain its positive impact on
vocal function. Reinjections could probably be accomplished as an outpatient procedure. This 3-month
outpatient treatment interval for chronic vocal fold
scarring is generally in line with the timing of the
repeated Botox injections that are currently used to
manage chronic laryngeal dystonias. Devising strategies to increase the residence time of PEG30 by
using chemical engineering and other approaches,
however, is also an important goal.
In summary, the results of this initial assessment
provide support for the continued development and
testing of PEG30 as a potential treatment for hoarseness resulting from deficiencies in pliability and
contour of the phonatory mucosa.
CONCLUSIONS
The PEG30 hydrogel is a promising candidate
biomaterial to restore vibration to scarred or deficient phonatory mucosa, since PEG30 exhibits in
vivo biocompatibility and pliability without impeding normal SLP function. These characteristics support a potential treatment paradigm for a majority
of cases of irresolvable hoarseness, including vocal
fold scar, since most patients have some residual pliable mucosa impeded by focal zones of stiffness.
REFERENCES
1. Brunings W. Technique of autoscopic operations. Direct laryngoscopy, bronchoscopy, and esophagoscopy. London:
Bailliere, Tindall, & Cox, 1912:118-20.
2. Payr E. Plastik am schildknorpel zur Behebung der Folgen einseitiger Stimmbandlahmung. Dtsch Med Wochenschr
1915;43:1265-70.
3. Arnold GE. Vocal rehabilitation of paralytic dysphonia.
I. Cartilage injection into a paralyzed vocal fold. AMA Arch
Otolaryngol 1955;62:1-17.
4. Isshiki N, Morita H, Okamura H, Hiramoto M. Thyroplasty as a new phonosurgical technique. Acta Otolaryngol
1974;78:451-7.
10. Allen J. Cause of vocal fold scar. Curr Opin Otolaryngol
Head Neck Surg (in press).
11. Bless DM, Welham NV. Characterization of vocal fold
scar formation, prophylaxis, and treatment using animal models. Curr Opin Otolaryngol Head Neck Surg (in press).
12. Chhetri DK, Mendelsohn AH. Hyaluronic acid for the
treatment of vocal fold scars. Curr Opin Otolaryngol Head
Neck Surg (in press).
13. Kishimoto Y, Hirano S, Kitani Y, et al. Chronic vocal
fold scar restoration with hepatocyte growth factor hydrogel.
Laryngoscope 2010;120:108-13.
5. Isshiki N, Tanabe M, Sawada M. Arytenoid adduction for
unilateral vocal cord paralysis. Arch Otolaryngol 1978;104:5558.
14. Kishimoto Y, Welham NV, Hirano S. Implantation of
atelocollagen sheet for vocal fold scar. Curr Opin Otolaryngol
Head Neck Surg (in press).
6. Zeitels SM, Hochman I, Hillman RE. Adduction arytenopexy: a new procedure for paralytic dysphonia with implications for implant medialization. Ann Otol Rhinol Laryngol
Suppl 1998;107(suppl 173):1-24.
15. Long JL. Tissue engineering for treatment of vocal fold
scar. Curr Opin Otolaryngol Head Neck Surg (in press).
7. Zeitels SM, Healy GB. Laryngology and phonosurgery.
N Engl J Med 2003;349:882-92.
8. Zeitels SM, Blitzer A, Hillman RE, Anderson RR. Foresight in laryngology and laryngeal surgery: a 2020 vision. Ann
Otol Rhinol Laryngol Suppl 2007;116(suppl 198):1-16.
9. Roy N, Merrill RM, Thibeault S, Parsa RA, Gray SD,
Smith EM. Prevalence of voice disorders in teachers and the
general population. J Speech Lang Hear Res 2004;47:281-93.
16. Sataloff RT. Autologous fat implantation for vocal fold
scar. Curr Opin Otolaryngol Head Neck Surg (in press).
17. Suehiro A, Hirano S, Kishimoto Y, Rousseau B, Nakamura T, Ito J. Treatment of acute vocal fold scar with local injection of basic fibroblast growth factor: a canine study. Acta
Otolaryngol 2010;130:844-50.
18. Molteni G, Bergamini G, Ricci-Maccarini A, et al. Autocrosslinked hyaluronan gel injections in phonosurgery. Otolaryngol Head Neck Surg 2010;142:547-53.
19. Finck C, Lefebvre P. Implantation of esterified hyaluronic
96
Karajanagi et al, Canine Vocal Fold Function After Injection of New Biomaterial
acid in microdissected Reinke’s space after vocal fold microsurgery: first clinical experiences. Laryngoscope 2005;115:18417.
20. Finck CL, Harmegnies B, Remacle A, Lefebvre P. Implantation of esterified hyaluronic acid in microdissected Reinke’s space after vocal fold microsurgery: short- and long-term
results. J Voice 2010;24:626-35.
21. Duflo S, Thibeault SL, Li W, Shu XZ, Prestwich GD. Vocal fold tissue repair in vivo using a synthetic extracellular matrix. Tissue Eng 2006;12:2171-80.
22. Hansen JK, Thibeault SL, Walsh JF, Shu XZ, Prestwich
GD. In vivo engineering of the vocal fold extracellular matrix
with injectable hyaluronic acid hydrogels: early effects on tissue repair and biomechanics in a rabbit model. Ann Otol Rhinol
Laryngol 2005;114:662-70.
23. Jahan-Parwar B, Chhetri DK, Ye M, Hart S, Berke GS.
Hylan B gel restores structure and function to laser-ablated canine vocal folds. Ann Otol Rhinol Laryngol 2008;117:703-7.
24. Hirano M. Phonosurgery: basic and clinical investigations. Otologia (Fukuoka) 1975;21:239-442.
25. Chan RW, Titze IR. Viscoelastic shear properties of human vocal fold mucosa: measurement methodology and empirical results. J Acoust Soc Am 1999;106:2008-21.
26. Zeitels SM, Vaughan CW. A submucosal true vocal fold
infusion needle. Otolaryngol Head Neck Surg 1991;105:478-9.
27. Mehta DD, Deliyski DD, Zeitels SM, Quatieri TF, Hillman RE. Voice production mechanisms following phonosurgical treatment of early glottic cancer. Ann Otol Rhinol Laryngol
2010;119:1-9.
28. Herrera VL, Viereck JC, Lopez-Guerra G, et al. 11.7 Tesla magnetic resonance microimaging of laryngeal tissue architecture. Laryngoscope 2009;119:2187-94.
Potential of Induced Pluripotent Stem Cells for the
Regeneration of the Tracheal Wall
Mitsuyoshi Imaizumi, MD; Yukio Nomoto, MD; Takashi Sugino, MD;
Masao Miyake, PhD; Ikuo Wada, PhD; Tatsuo Nakamura, MD; Koichi Omori, MD
Objectives: Our previous studies focused on basic research and the clinical applications of an artificial trachea. However,
the prefabricated artificial trachea cannot be utilized for pediatric airways, because the tracheal frame needs to expand as
the child develops. The purpose of this study was to evaluate the potential of induced pluripotent stem (iPS) cells for the
regeneration of the tracheal wall.
Methods: We cultured iPS cells in a 3-dimensional (3-D) scaffold in chondrocyte differentiation medium (bioengineered
scaffold model), and the results were compared with those in a 3-D scaffold without iPS cells (control scaffold model).
The 3-D scaffolds were implanted into tracheal defects in 8 nude rats. After 4 weeks, the regenerated tissue was histologically examined.
Results: Implanted iPS cells were confirmed to exist in all 5 rats implanted with bioengineered scaffolds. Cartilage-like
tissue was observed in the regenerated tracheal wall in 2 of the 5 rats in the bioengineered scaffold model, but in none of
the 3 rats in the control scaffold model.
Conclusions: Implanted iPS cells were confirmed to exist in the bioengineered scaffold. Cartilage-like tissue was regenerated in the tracheal defect. This study demonstrated the potential of iPS cells in the regeneration of the tracheal wall.
Key Words: bioengineered scaffold, chondrogenesis, induced pluripotent stem cell, regeneration, tracheal wall.
the regeneration of tracheal tissue.3-5 This technique
was used in 4 patients,6 and all cases have shown
good progress without restenosis. However, the premade artificial trachea cannot be utilized for pediatric airways, because the tracheal frame needs to
expand as the child develops. Even now, the treatment of pediatric laryngotracheal stenosis remains a
difficult challenge, because treatment often requires
multistaged procedures and successful decannulation sometimes fails after a series of operations.7,8
INTRODUCTION
Reconstruction of the upper airway after the resection of malignancies or stenotic inflammatory lesions is thought to be difficult. The trachea is composed of the tracheal epithelium, a ligament comprising collagen fibers for the maintenance of elasticity, and cartilage. Ideally, both the airway framework and the endotracheal surface should be reconstructed. Several types of flaps and grafts, made
from cartilage, muscle, skin, and so on, have been
used in the repair of defects after the resection of
tracheal lesions.1,2 However, postoperative scarring or granulation is sometimes observed, leading
to airway stenosis. Therefore, reconstruction of the
respiratory tract with full functionality is vital after
surgical treatment for cancer or stenotic inflammatory lesions. Our group has developed and clinically applied an artificial trachea made from a collagen sponge with a polypropylene scaffold frame for
If the tracheal wall could be regenerated with patient-derived cells, this problem would be solved.
Previously, novel embryonic stem (ES) cell–like
pluripotent cells, known as induced pluripotent stem
(iPS) cells, were generated from mouse skin fibroblasts by the introduction of 4 transcription factors.9
These cells are capable of unlimited symmetric selfrenewal, thus providing an unlimited cell source for
tissue engineering applications. In addition, the use
From the Department of Otolaryngology (Imaizumi, Nomoto, Omori), the Department of Basic Pathology (Sugino), the First Department of Physiology (Miyake), and the Department of Cell Science, Institute of Biomedical Sciences (Wada), School of Medicine,
Fukushima Medical University, Fukushima City, and the Department of Bioartificial Organs, Institute for Frontier Medical Sciences,
Kyoto University, Kyoto (Nakamura), Japan. This study was supported in part by a Challenging Exploratory Research grant from the
Japan Society for the Promotion of Science and in part by a Grant-in-Aid for Young Scientists (Startup) from the Japan Society for the
Promotion of Science.
Presented at the meeting of the American Broncho-Esophagological Association, Las Vegas, Nevada, April 28-29, 2010.
Correspondence: Mitsuyoshi Imaizumi, MD, Dept of Otolaryngology, Fukushima Medical University, School of Medicine, 1 Hikarigaoka, Fukushima 960-1295, Japan.
97
98
Imaizumi et al, Induced Pluripotent Stem Cells
ated cells.14
For this purpose, we prepared puromycin-resistant
SNL 76/7 cells. Briefly, the Streptomyces alboniger
puromycin-N-acetyl-transferase gene was subcloned
into a retrovirus shuttle vector, pCX4bsr,15 and introduced into SNL 76/7 cells by infecting the recombinant retrovirus vector as described previously.16 The
puromycin-resistant SNL cells were selected by culturing in DMEM in the presence of 2 μmol/L puromycin.
Fig 1. Green fluorescent protein–expressing undifferentiated induced pluripotent stem (iPS) cell colonies. Bar
— 100 μm.
of iPS cells obtained from patient-derived somatic
cells can prevent transplant rejection. Recent studies
have found that iPS cells could provide a new cell
source for regeneration therapy.10-13
The purpose of this study was to evaluate the potential of iPS cells in the regeneration of the tracheal wall through the effective use of a bioengineered
scaffold containing iPS cells.
MATERIALS AND METHODS
Cell Culture. Induced pluripotent stem cells from
the iPS-MEF-Ng-20D-17 line (20D-17 purchased
from Riken BioResource Center, Tsukuba, Japan)
were routinely cultured on a feeder layer of mitomycin C–inactivated mouse embryonic fibroblasts,
SNL 76/7 (DS Pharma Biomedical Co, Ltd, Osaka,
Japan) in a cultivation medium consisting of Dulbecco’s modified Eagle’s medium (DMEM; Gibco,
Invitrogen, Grand Island, New York) supplemented with 15% fetal bovine serum (FBS; SAFC Biosciences, Lenexa, Kansas), 2 mmol/L L-glutamine
(Gibco, Invitrogen), 0.4 mL β-mercaptoethanol
(Sigma-Aldrich, St Louis, Missouri), and nonessential amino acids (Gibco, Invitrogen). Induced pluripotent stem cell lines were passaged every second
day with 0.25% trypsin–ethylenediaminetetraacetic
acid (Gibco, Invitrogen). To maintain iPS cell pluripotency, we cultured iPS cells in the above-mentioned medium on puromycin-resistant SNL 76/7
cells containing 1.0 μg/mL puromycin (Sigma-Aldrich) during selection of undifferentiated cells, to
select green fluorescent protein (GFP)–expressing
undifferentiated cells (Fig 1), on a gelatin-coated
dish (6 cm in diameter). The GFP expression became negative when differentiation was induced,
whereas GFP expression remained in undifferenti-
Morphological Examination In Vitro. Induced
pluripotent stem cells were cultured on a gelatincoated dish in a chondrocyte differentiation medium (CDM; Cell Applications, Inc, San Diego, California) to confirm chondrogenic differentiation of
iPS cells. As a control, iPS cells were cultured on
a gelatin-coated dish in 10% FBS–DMEM. After 6
weeks of cultivation, iPS cells were stained with Alcian blue to detect acidic proteoglycans produced by
chondrocytes.17
Detection of Gene Expression by Quantitative
Reverse Transcription–Polymerase Chain Reaction.
Total RNA was isolated with an RNeasy Mini-Kit
(Qiagen, Hilden, Germany). Samples were treated with Turbo DNase-free (Ambion, Austin, Texas) to remove traces of genomic DNA contamination. Total RNA (0.4 μg) was reverse-transcribed
to complementary DNA by use of Superscript III
Reverse Transcriptase (Invitrogen, Carlsbad, California) and oligo-dT primer according to the manufacturer’s instructions. Real-time polymerase chain
reaction was performed with master mixes (Applied Biosystems, Foster City, California) for SYBR
green incorporation and an ABI Prism 7300 detector. The thermal cycling conditions were 50°C for
2 minutes and 95°C for 10 minutes, followed by
40 cycles of 95°C for 15 seconds and 60°C for 1
minute. The following premade primers purchased
from TaKaRa Bio (Shiga, Japan) were used: type
II collagen (Col2a1; MA067811), type I collagen
(Col1a1; MA093358), and glyceraldehyde-3-phosphate dehydrogenase (GAPDH; MA050371). The
primer sequence for genes encoding the matrix
protein type II collagen was 5'-AGGGCAACAGCAGGTTCACATAC-3', 5'-TGTCCACACCAAATTCCTGTTCA-3'. Bone matrix protein collagen
I (5'-ATGCCGCGACCTCAAGATG-3', 5'-TGAGGCACAGACGGCTGAGTA-3') and the “housekeeping gene” GAPDH (5'-TGTGTCCGTCGTGGATCTGA-3', 5'-TTGCTGTTGAAGTCGCAGGAG-3') were used as internal standards.
Fabrication of Bioengineered Scaffold and Differentiation of iPS Cells. Omori et al5 and Nakamura
Imaizumi et al, Induced Pluripotent Stem Cells
99
Fig 2. Fabrication of bioengineered scaffold.
Three-dimensional (3-D) scaffold consists of collagenous sponge and polypropylene mesh. Control scaffold model is 3-D scaffold with gel only.
Bioengineered scaffold model is 3-D scaffold with
gel containing chondrogenic cells differentiated
from iPS cells. CDM — chondrocyte differentiation medium.
et al3 developed an artificial trachea made from a
collagen sponge scaffold and polypropylene mesh
based on the concept of in situ tissue engineering for
tracheal regeneration. The polypropylene mesh used
was a conventional material developed for clinical
applications, and the collagen sponge consisted of
type I and type III collagens of porcine origin. The
procedure for the production of the artificial trachea
was described by Nakamura et al.3 The sponge, with
a pore size range of 100 to 500 μm, was made from
porcine dermal atelocollagen, consisting of type I
(70% to 80%) and type III collagen. The atelocollagen was dissolved in a hydrochloric acid solution, pH 3.0, at a concentration of 1.2%, stirred at
8,000 rpm for 15 minutes, freeze-dried, and heated
at 140°C in a vacuum for 24 hours. In our current
study, we used a 3-dimensional (3-D) scaffold (diameter, 10 mm; thickness, 3 mm) made from a piece
of artificial trachea.
Collagen gel containing iPS cells was prepared
as follows. A type I collagen solution (Nitta Gelatin,
Osaka), fivefold-concentrated DMEM/F-12, and reconstituted buffer (25 mmol/L Hepes, 0.15 mol/L
sodium hydroxide) were mixed at a ratio of 7:2:1.
Induced pluripotent stem cells were suspended in
the reconstituted collagen solution at a density of
5.0 × 106 cells per milliliter. To cultivate the iPS
cells, we used a 3-D scaffold made from the abovementioned collagen sponge scaffold with polypropylene mesh. Four hundred microliters of 0.3% collagenous solution containing the iPS cells was introduced into the 3-D scaffold (2.0 × 106 cells per
scaffold) in Millicell Culture Plate inserts (Millipore
Co, Billerica, Massachusetts) with a membrane filter (0.4 μm in pore size), and the collagenous solution was allowed to infiltrate the scaffold. The bioengineered scaffolds were then incubated at 37°C
for at least 30 minutes, and the collagenous solution
formed a gel. These bioengineered scaffolds were
placed in 6-well polystyrene plates (Becton Dickinson, Franklin Lakes, New Jersey) containing 2 mL
of CDM (bioengineered scaffold model). As a control, collagen gel without iPS cells (control scaffold
model) infiltrated into a 3-D scaffold was cultured
in 10% FBS–DMEM. After 6 weeks of cultivation
in vitro, each of the scaffolds was implanted into
nude mice (Fig 2).
Implantation Into Tracheal Defects in Rats. This
study was carried out under the control of the Animal Care and Use Committee in accordance with
Guidelines for Animal Experiments of Fukushima
Medical University, the Japanese Government Law
Concerning the Protection and Control of Animals,
and the Japanese Government Notification on Feeding and Safekeeping of Animals.
A total of eight 8- to 10-week-old, male F344/
NJcl-rnu/rnu nude rats were used as implant recipients in this study. The 8 rats each received one of the
two types of 3-D scaffold (bioengineered scaffold
or control scaffold). Tracheostomy and implantation
of one of the two types of scaffold were performed
under general anesthesia induced by inhalation of
isoflurane. The cervical tracheas were exposed
through a vertical skin incision, and the sternohyoid
and sternothyroid muscles were split along the median line. Tracheal defects, approximately 1.5 mm
in width by 2.5 mm in length, were then formed.
The scaffolds were laid onto the tracheal defects. In
order to prevent shifting of the graft, we then replaced the sternohyoid and sternothyroid muscles
over the graft and sutured them. Finally, the incised
skin was sutured. At 28 days after the operation, the
rats were painlessly sacrificed, and the tracheas and
the sternohyoid and sternothyroid muscles were removed en bloc (Fig 2). The samples were fixed in
100
Imaizumi et al, Induced Pluripotent Stem Cells
A
B
Fig 3. Morphological examination in vitro. Bars — 100 μm. A) Six weeks after cultivation, iPS cells cultured in CDM show
Alcian blue–stained areas around iPS cell colonies. B) In contrast, iPS cells cultured in 10% fetal bovine serum–Dulbecco’s
modified Eagle’s medium show no Alcian blue–stained areas.
4% paraformaldehyde, embedded in paraffin, and
sliced for hematoxylin and eosin staining and immunohistochemical analysis. To detect differentiation of the tissue components, we performed immunohistochemical analysis using antibodies to cytokeratin AE1/3 (DAKO, Tokyo, Japan) for the epithelium, glial fibrillary acidic protein (DAKO) for the
nervous system, and muscle-specific actin (Enzo,
New York, New York) for muscles.
RESULTS
Morphological Examination In Vitro. Six weeks
after cultivation, iPS cells cultured in CDM showed
Alcian blue–stained areas around iPS cell colonies
(Fig 3A). In contrast, iPS cells cultured in 10%
FBS–DMEM showed no Alcian blue–stained areas
(Fig 3B).
Detection of Gene Expression by Quantitative
RT-PCR Analysis. Messenger RNA expression of
type II collagen was increased 4 weeks after cultivation with CDM. Expression of type I collagen, an
osteogenic differentiation marker, was not detected
in either group at any time point (Fig 4).
Implantation of 3-D Scaffolds Into Tracheal Defects in Rats. Survival of the iPS cells was confirmed
in the bioengineered scaffold in all 5 rats, from the
formation of various tissues derived from the three
germ layers of the teratoma. Glands, nerves, and
muscles were identified by hematoxylin and eosin
staining and immunohistochemical analysis (Fig 5).
The teratomas were identified just outside the implants in all cases.
In the regenerated tissue of the tracheal wall, cartilage-like tissue was observed (Fig 6) in 2 of the 5
rats in the bioengineered scaffold model. One and
two cartilage-like tissue foci, respectively, were
identified in the 2 rats. The cartilage-like formations
identified were one tenth the size of the cartilage defect in the original trachea. On the other hand, no
cartilage-like tissue was observed in any of the 3
rats in the control scaffold group (Fig 7).
DISCUSSION
Cartilage regeneration therapy has been applied
clinically to the treatment of osteoarthritis. The first
implantation of autologous cultured chondrocytes
Fig 4. Polymerase chain reaction–
based analysis of chondrocyte differentiation in vitro. Messenger
RNA expression of type II collagen was increased 4 weeks after
cultivation. Expression of type I
collagen, osteogenic differentiation marker, was not detected in
either group at any time point. A)
Type II collagen. B) Type I collagen.
A
B
Imaizumi et al, Induced Pluripotent Stem Cells
101
Fig 5. Histologic findings of teratoma formation in vivo. In bioengineered scaffold, tissues associated with three germ layers of teratoma,
such as glands (endoderm), nerves (ectoderm), and muscles (mesoderm), were identified by B,D,F) hematoxylin and eosin staining and
C,E,G) immunohistochemical analysis performed with antibodies to
cytokeratin AE1/3 (C), glial fibrillary acidic protein (E), and musclespecific actin (G). A) Original ×40. Bar — 1 mm; arrows — teratoma formation in bioengineered scaffold. B,C) Glands (original ×400).
D,E) Nerves (original ×400). F,G) Muscles (original ×400).
A
B
D
F
C
E
G
was performed in the treatment of knees with articular cartilage defects more than 20 years ago. To
reduce surgical invasiveness and complications, the
procedure for chondrocyte implantation has been improved; however, no consensus has been reached regarding this treatment.18 Further difficulties remain,
in that the amount of autologous chondrocytes is
limited, and invasive procedures are essential. Thus,
numerous studies have focused on the differentiation of chondrogenic cells in vitro, with mesenchymal cell lines and ES cell lines used as potential cell
sources.19-24 These reports show that mesenchymal
stem cells and ES cells have chondrogenic potential.
Although mesenchymal cell lines, such as mesenchymal stem cells from bone marrow, are relatively
safe, these cells have limited ability to proliferate
and differentiate, and their ability to do so is further decreased after several passages. On the other
hand, ES cells are capable of unlimited proliferation
and differentiation into any tissue or cell. However, teratoma formation and ethical issues have posed
problems for tissue engineering applications using
ES cells.
This is the first demonstration of iPS-derived cells
differentiating into chondrocytes in a 3-D scaffold
for tracheal regeneration. Induced pluripotent stem
cells have the potential to differentiate into chondrogenic cells, as shown by the appearance of Alcian blue–stained colonies and expression of a cartilage-associated gene. Normal iPS cells were slightly
stained with Alcian blue, whereas iPS cell colonies
cultured with CDM were strongly stained with Alcian blue. This finding suggests that proteoglycan is
strongly expressed in differentiated iPS cells.
This study suggests the potential for iPS cells to
be used as a new cell source in tracheal regeneration
therapy.
Our results show that iPS cells differentiate into
chondrocytes in vitro. Cartilage-like tissue was observed in vivo in the 3-D scaffold containing differ-
102
Imaizumi et al, Induced Pluripotent Stem Cells
A
A
B
B
Fig 6. Histologic findings of implanted bioengineered
scaffold in vivo. A) Original ×40. Bar — 1 mm. B) Original ×400. Bar — 100 μm. Cartilage-like tissue was observed in rats in bioengineered scaffold model.
Fig 7. Histologic findings of implanted control scaffold
in vivo. A) Original ×40. Bar — 1 mm. B) Original ×400.
Bar — 100 μm. No cartilage tissue was observed in rats
in control scaffold model.
entiated iPS cells. In our current study, we implanted
the 3-D scaffold containing differentiated iPS cells
after culture for 6 weeks into in vivo tracheal defects
because most of the chondrocytic cells were stained
with Alcian blue in vitro.
release of various factors appropriate to maintain a
chondrogenic phenotype. However, the regenerated
tissue was rather far from the area of the tracheal
cartilage defects. In future, we intend to further investigate scaffold size, thickness, and gel volume to
better match the tracheal defect area.
In this study, cartilage-like tissue could be confirmed in vivo. However, similar studies using ES
cells failed to observe cartilage tissue formation in
vivo, although histologic and polymerase chain reaction analysis has demonstrated chondrogenic differentiation in vitro.24,25 The reason suggested for this
finding was that predifferentiation in vitro was insufficient to maintain a stable chondrogenic phenotype in vivo. Grafts, in these studies, were implanted
ectopically in the subcutaneous tissue of the backs
of nude mice. The lack of chondrogenic signals in
the ectopic implantation site suggests the dedifferentiation or cell death of the implanted cartilaginous
tissue. Thus, continuous chondrogenic stimulation
may need to be maintained. In our study, differentiated iPS cells were implanted in tracheal cartilage
defects. The conditions in this case may induce the
In the bioengineered scaffold, the survival of
iPS cells was confirmed by teratoma formation. In
this study, all implanted iPS cells formed teratomas. There are no previous reports on the use of iPS
cells for tracheal cartilage regeneration; however,
the current study has indicated that teratoma formation from iPS cells may be affected by the method
used for reprogramming and differentiation, the site
of transplantation, the host background, and other
factors. It was also shown that there was a significant correlation between teratoma size and undifferentiated iPS cell content.26 We tried to induce the
differentiation of the iPS cells to chondrocytes using bone morphogenetic protein 2. However, these
iPS cells also formed teratomas (data not shown).
To reduce teratoma formation, we will attempt to re-
Imaizumi et al, Induced Pluripotent Stem Cells
duce the number of undifferentiated cells by using
more effective methods that were described previously.13,22,23 Information with regard to the ratio of
differentiated to undifferentiated cells would also
be useful in dealing with the problem of teratoma
formation. Further studies on inducing the effective
differentiation of iPS cells to chondrocytes in vitro
are expected.
CONCLUSIONS
This is the first demonstration that iPS-derived
103
cells differentiate into chondrocytes in 3-D scaffolds implanted for tracheal regeneration. Our study
demonstrated that iPS cells have the potential to differentiate into chondrogenic cells, as shown by the
appearance of Alcian blue–stained colonies and expression of a cartilage-associated gene of collagen
type II in vitro. Implanted iPS cells were confirmed
to exist in the 3-D scaffold in vivo. Cartilage-like
tissue was regenerated in tracheal defects with use
of iPS cells. Our results suggest the potential of iPS
cells in the regeneration of the tracheal wall.
Acknowledgments: We thank Etsuko Sato and Yuka Sato for their technical assistance.
REFERENCES
1. Caputo V, Consiglio V. The use of patient’s own auricular
cartilage to repair deficiency of the tracheal wall. J Thorac Cardiovasc Surg 1961;41:594-6.
2. Cotton RT. Management of subglottic stenosis. Otolaryngol Clin North Am 2000;33:111-30.
3. Nakamura T, Teramachi M, Sekine T, et al. Artificial trachea and long-term follow-up in carinal reconstruction in dogs.
Int J Artif Organs 2000;23:718-24.
4. Kawaguchi S, Nakamura T, Shimizu Y, et al. Mechanical properties of artificial tracheas composed of a mesh cylinder
and a spiral stent. Biomaterials 2001;22:3085-90.
5. Omori K, Nakamura T, Kanemaru S, et al. Regenerative
medicine of the trachea: the first human case. Ann Otol Rhinol
Laryngol 2005;114:429-33.
6. Omori K, Tada Y, Suzuki T, et al. Clinical application of
in situ tissue engineering using a scaffolding technique for reconstruction of the larynx and trachea. Ann Otol Rhinol Laryngol 2008;117:673-8.
7. Hartley BE, Cotton RT. Paediatric airway stenosis: laryngotracheal reconstruction or cricotracheal resection? Clin Otolaryngol Allied Sci 2000;25:342-9.
8. Gustafson LM, Hartley BE, Cotton RT. Acquired total
(grade 4) subglottic stenosis in children. Ann Otol Rhinol Laryngol 2001;110:16-9.
9. Takahashi K, Yamanaka S. Induction of pluripotent stem
cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell 2006;126:663-76.
10. Karumbayaram S, Novitch BG, Patterson M, et al. Directed differentiation of human-induced pluripotent stem cells
generates active motor neurons. Stem Cells 2009;27:806-11.
11. Senju S, Haruta M, Matsunaga Y, et al. Characterization
of dendritic cells and macrophages generated by directed differentiation from mouse induced pluripotent stem cells. Stem Cells
2009;27:1021-31.
12. Tashiro K, Inamura M, Kawabata K, et al. Efficient adipocyte and osteoblast differentiation from mouse induced pluripotent stem cells by adenoviral transduction. Stem Cells 2009;
27:1802-11.
13. Taura D, Sone M, Homma K, et al. Induction and isolation of vascular cells from human induced pluripotent stem cells
— brief report. Arterioscler Thromb Vasc Biol 2009;29:11003.
14. Okita K, Ichisaka T, Yamanaka S. Generation of germline-competent induced pluripotent stem cells. Nature 2007;
448:313-7.
15. Akagi T, Sasai K, Hanafusa H. Refractory nature of normal human diploid fibroblasts with respect to oncogene-mediated transformation. Proc Natl Acad Sci U S A 2003;100:1356772.
16. Hatsuzawa K, Tamura T, Hashimoto H, et al. Involvement of syntaxin 18, an endoplasmic reticulum (ER)–localized
SNARE protein, in ER-mediated phagocytosis. Mol Biol Cell
2006;17:3964-77.
17. Steedman HF. Alcian blue 8GS: a new stain for mucin. J
Cell Sci 1950;1:477-9.
18. Brittberg M. Cell carriers as the next generation of cell
therapy for cartilage repair: a review of the matrix-induced autologous chondrocyte implantation procedure. Am J Sports Med
2010;38:1259-71.
19. Ahrens M, Ankenbauer T, Schröder D, Hollnagel A, Mayer H, Gross G. Expression of human bone morphogenetic proteins–2 or –4 in murine mesenchymal progenitor C3H10T1/2
cells induces differentiation into distinct mesenchymal cell lineages. DNA Cell Biol 1993;12:871-80.
20. Mackay AM, Beck SC, Murphy JM, Barry FP, Chichester CO, Pittenger MF. Chondrogenic differentiation of cultured human mesenchymal stem cells from marrow. Tissue Eng
1998;4:415-28.
21. Solchaga LA, Penick K, Goldberg VM, Caplan AI, Welter JF. Fibroblast growth factor–2 enhances proliferation and
delays loss of chondrogenic potential in human adult bone-marrow–derived mesenchymal stem cells. Tissue Eng Part A 2010;
16:1009-19.
22. Kramer J, Hegert C, Guan K, Wobus AM, Müller PK,
Rohwedel J. Embryonic stem cell–derived chondrogenic differentiation in vitro: activation by BMP-2 and BMP-4. Mech Dev
2000;92:193-205.
23. Wakitani S, Aoki H, Harada Y, et al. Embryonic stem
cells form articular cartilage, not teratomas, in osteochondral
defects of rat joints. Cell Transplant 2004;13:331-6.
24. Jukes JM, Moroni L, Van Blitterswijk CA, de Boer J.
Critical steps toward a tissue-engineered cartilage implant using embryonic stem cells. Tissue Eng Part A 2008;14:135-47.
25. De Bari C, Dell’accio F, Luyten FP. Failure of in vitro–
differentiated mesenchymal stem cells from the synovial membrane to form ectopic stable cartilage in vivo. Arthritis Rheum
2004;50:142-50.
26. Miura K, Okada Y, Aoi T, et al. Variation in the safety
of induced pluripotent stem cell lines. Nat Biotechnol 2009;27:
743-5.
Determination of Predictive Factors of Tracheobronchial
Prosthesis Removal: Stent Brands Are Crucial
Guillaume Buiret, MD; Carole Colin, MD; Guillaume Landry, MD;
Marc Poupart, MD; Jean-Christian Pignat, MD
Objectives: We sought to describe in a retrospective study our experience in the endoscopic management of tracheobronchial stenoses over 20 years and to determine prognostic factors of stent removal.
Methods: We analyzed the medical records of 166 patients (111 male and 55 female) who underwent the placement of a
prosthesis for all causes of tracheobronchial stenosis between 1990 and 2009.
Results: Overall, 34% of the patients had their stents removed. The incidence of complications for the first stent was 0.08
per patient-month. One hundred five patients (63%) had no complications. In univariate analysis, stent removal was significantly linked with the stent brand. In multivariate analysis, taking into account the causes of stenosis, the stent brand
appeared to be the only factor that significantly influenced stent removal. Finally, stenosis with more than 1 stent replacement was most prone to repeat endoscopies.
Conclusions: Even though endoscopic stent placement is a relatively safe and effective treatment for tracheobronchial
stenoses, particularly in cases with malignancy, complications led to stent removal in about one third of cases. The type
of stent chosen is crucial.
Key Words: bronchoscopy, stent, upper airway.
INTRODUCTION
fore TBS placement.
The complications of TBSs are frequent and often
require repeated endoscopies that sometimes lead to
removal of the TBS. The aim of this study was to
evaluate the predictive factors for TBS removal in
order to warn practitioners about the risks of TBSs.
Although tracheobronchial stents (TBSs) are not
recent,1 they have only been used routinely since
the end of the 1990s. Their purpose is to restore
the tracheobronchial patency reduced by endoluminal cellular proliferation, external compression, or
both,2 often while patients are waiting for another
treatment (such as chemotherapy and/or radiotherapy3,4). Placement may be preceded by a preparatory treatment.5 A formal contraindication to surgical treatment (related to the disease or the general
status of the patient) leaves two indications for TBS
placement: 1) malignancies (obstruction by extrinsic and/or intrinsic compression, tumoral progression in spite of frequent laser treatments, tracheoesophageal fistula [TEF], postobstructive pneumonia6); and 2) benign causes (pulmonary transplantation, prolonged intubation, tracheotomy, irradiation,
polychondritis, congenital and idiopathic granulomatosis,7 loss of cartilaginous support6). However,
the decision not to operate must be made in expert
centers. In the series of Gaissert et al,8 for example,
two thirds of the cases were considered operable be-
MATERIALS AND METHODS
Patient Selection. A list of the patients treated between January 1, 1990, and September 30, 2009,
for TBS placement in the Otorhinolaryngology and
Cervicofacial Surgery Unit of the Croix-Rousse
University Hospital was provided by the hospital’s
Department of Medical Information. The data were
compared to the records of the endoscopy room, in
which all of the results of endoscopies and interventional endoscopies are systematically indexed. The
files of 166 patients were finally retained and retrospectively analyzed.
Method of Treatment. The patients underwent
placement of a TBS, possibly preceded by bronchial
de-obstruction (with forceps and/or a laser), during
From the Otorhinolaryngology and Cervicofacial Surgery Unit, Hôpital de la Croix-Rousse, Centre Hospitalier Universitaire Lyon (all
authors), and the Université Claude Bernard Lyon 1 (Buiret, Colin, Landry, Pignat), Lyon, France.
Presented at the meeting of the American Broncho-Esophagological Association, Las Vegas, Nevada, April 28-29, 2010.
Correspondence: Guillaume Buiret, MD, Otorhinolaryngology and Cervicofacial Surgery Unit, Hôpital de la Croix-Rousse, 104
Grande Rue de la Croix-Rousse, 69004 Lyon, France.
104
Buiret et al, Is Stent Brand Predictive Factor of Complications?
105
TABLE 1. CHARACTERISTICS OF POPULATION
Sex
Male
Female
Mean age
Age range
Causes of stenosis
Benign
Extrinsic compression
Tracheoesophageal fistula
Pulmonary transplantation
Intubation
Tracheal resection-anastomosis
Tracheomalacia
Tracheotomy
Two or more causes
Others
Malignant
Extrinsic compression
Tracheoesophageal fistula
Lung cancer
Esophageal cancer
Thyroid cancer
Tracheal cancer
Others
111 (67%)
55 (33%)
59.7 y (SE, 16.1 y)
6.6 to 90.1 y
91 (55%)
8
4
12
14
5
9
28
4
7
75 (45%)
35
19
9
2
3
3
4
tracheobronchial endoscopy with suspension laryngoscopy (method described previously9). Several
brands of prostheses were used during the study period.
The following nonexpandable prostheses were
used: Aboulker (10 prostheses placed between 1981
and 2005), Cook (William Cook Europe, Bjaeverskov, Denmark; 8 prostheses placed between 1990
and 1996), Novatech Y (Novatech SA, La Ciotat,
France; 24 prostheses placed between 2002 and
2009), and Rush Y (Boston Scientific, Natick, Massachusetts; 23 prostheses placed between 2001 and
2008).
The following expandable covered prostheses
were used: Wallstent (Boston Scientific; 20 prostheses placed between 1997 and 2008), Hanarostent
(MI Tech Co, Pyeongtaek, South Korea; 6 prostheses placed between 2002 and 2005), Novatech Silmet (Novadis, Holy Victoret, France; 25 prostheses
placed between 2007 and 2009), Novatech Y (Novadis; 1 prosthesis placed in 2009), Tracheobronxane (Novatech SA; 19 prostheses placed between
2001 and 2007), and Ultraflex (Boston Scientific;
15 prostheses placed between 1981 and 2005).
The only expandable uncovered prosthesis used
was the Wallstent (Boston Scientific; 10 prostheses
placed between 1994 and 2005).
End Point. The aim of the study was to establish
Fig 1. Overall survival by cause of stenosis.
predictive factors for removal of the TBS. The duration of prosthesis use was from the day of placement
to the time of death or to the date of the last followup visit, limited to September 30, 2009, for the remaining patients.
Data Analysis. Univariate analyses were performed, and statistical differences in the duration of
the prostheses were tested by a Cox model for a risk
α of 0.05. In case of significant predictive factors,
multivariate analyses taking into account the principal potential confounding factors were performed.
The statistical analyses were carried out with SPSS
v12.0 software.
RESULTS
The characteristics of the population are described
in Table 1, and Fig 1 presents the overall survival
curve of the patients.
The causes of stenosis were mainly benign (55%).
Nearly 60% of the benign stenoses were caused by
tracheomalacia, tracheotomy, or intubation. The 7
other benign causes (Table 1) comprised 1 tracheal
burn, 1 tracheal chondroma, 1 Lortat-Jacob mucosynechiant dermatitis, 1 Riedel’s thyroiditis, 1 tracheal radionecrosis, and 2 pseudo-inflammatory tumors.
Malignancies were the cause of stenosis in approximately 45% of the cases, and more than 75%
of these were intrinsic or extrinsic tumoral compressions. The 4 other malignant causes (Table 1) comprised 2 non-Hodgkin’s lymphomas, 1 Hodgkin’s
lymphoma, and 1 tracheal resection-anastomosis for
tracheal cancer.
The distribution of types of first prosthesis used is
presented in Table 2. Ninety-three expandable covered prostheses (56%), 12 expandable uncovered
prostheses (7%), and 61 nonexpandable prostheses
(37%) were used in the first intention. More than
65% of the stenoses with benign causes were treat-
106
Buiret et al, Is Stent Brand Predictive Factor of Complications?
TABLE 2. DISTRIBUTION OF TYPES OF FIRST PROSTHESIS USED BY CAUSE OF STENOSIS
Prosthesis Type
Cause
Benign
Extrinsic compression
Tracheoesophageal fistula
Pulmonary transplantation
Intubation
Tracheal resection and anastomosis
Tracheomalacia
Tracheotomy
Two or more causes
Others
Malignant
Extrinsic compression
Tracheoesophageal fistula
Lung cancer
Esophageal cancer
Thyroid cancer
Tracheal cancer
Others
Total
Expandable Covered
Expandable Uncovered
Nonexpandable
61
4
3
6
12
4
4
22
2
4
32
9
9
9
0
3
2
0
93
10
1
0
2
1
0
2
4
0
0
2
2
0
0
0
0
0
0
12
20
3
1
4
1
1
3
2
2
3
41
24
10
0
2
0
1
4
61
ed with a covered expandable prosthesis. Approximately 40% and 50% of the stenoses with malignant
causes were treated with expandable covered and
nonexpandable prostheses, respectively.
The complication rate for the first prosthesis was
0.08 per patient-month (147 endoscopies for complications in 1,796 months of prosthesis use). One
hundred five patients (63%) did not have any complications.
The prostheses of 57 patients (34%) had to be removed: 3 on the day of placement, 1 on the 2nd day,
4 between the 3rd and 7th days, and 13 between the
8th and 30th days. Table 3 presents the reasons for
removal according to the type of prosthesis. Other
reasons for TBS removal comprised 1 failure of installation, 1 prosthesis fatigue, and 1 case of esophageal prosthesis placement, which made the TBS
unbearable. Figure 2 shows the distribution of stent
removal according to time. The mean duration before removal was 277 days (SE, 540 days), the median was 67 days, and the range was 0 days to 7.5
years.
Forty-five prostheses had to be replaced (79% of
the removed prostheses). The distribution of second
prosthesis types is presented in Table 3. Generally
speaking, when the first prosthesis was expandable
and covered, then the replacement prosthesis was
expandable and covered (66%) or nonexpandable
(34%). If the first prosthesis was nonexpandable,
then the replacement prosthesis usually was also
nonexpandable (85%).
Total
8
4
12
14
5
9
28
4
7
35
19
9
2
3
3
4
166
The analysis of the presumed predictive factors
for removal is presented in Table 4. The hazard ratios represent the probability of maintenance of the
prosthesis. The greater the ratio, the longer the prosthesis remained in place. Multivariate analysis made
it possible to take into account the causes and potential confounding factors.
The second prosthesis had to be removed in 18
patients (40%), and 17 of them had a third prosthesis. The third prosthesis had to be removed in 7 patients (39%), and 5 of them had a fourth prosthesis.
The fourth prosthesis had to be removed in 4 patients (57%), and 3 of them had a fifth prosthesis.
The fifth prosthesis had to be removed in 1 patient
(33%), who had a sixth prosthesis.
DISCUSSION
Our study on a broad series with several TBS
brands is the first to show that the only significant
predictive factor for prosthesis removal is the prosthesis brand used. Contrary to the study of Saad et
al,9 this finding was independent of the benign or
malignant nature and precise cause of the stenosis
and independent of the type of prosthesis (nonexpandable, expandable covered, or uncovered). In ascending order, from the least stable to the most stable, the prostheses were as follows: Ultraflex, covered Wallstent, Cook, Hanarostent, Aboulker, Tracheobronxane, Novatech Silmet, uncovered Wallstent, Novatech Y, and Rush Y.
The majority of prosthesis removals were per-
Buiret et al, Is Stent Brand Predictive Factor of Complications?
107
TABLE 3. REASONS FOR REMOVAL BY TYPE OF PROSTHESIS AND DISTRIBUTION OF SECOND PROSTHESIS TYPES
Reason for Removal
Migration
Granuloma
Sputum retention
No. of
Patients
19
13
6
Type of Prosthesis
No. of Patients/
Total No. of Stents
Expandable covered
13/93
Nonexpandable
6/60
Expandable covered
9/93
Expandable uncovered
Nonexpandable
1/12
3/60
Expandable covered
2/93
Nonexpandable
4/60
Infection
5
Expandable covered
4/93
Extremity overgrowth
4
Nonexpandable
Expandable covered
1/60
3/93
Ineffectiveness
3
Cough
2
Nonexpandable
Expandable covered
Expandable uncovered
Nonexpandable
Nonexpandable
1/60
1/93
1/12
1/60
2/60
Cure
Other reasons
2
3
Expandable covered
Expandable covered
Nonexpandable
2/93
2/93
1/60
formed within 1 year of implantation and in a quasisuperposable way, whether the cause of stenosis was
benign or malignant (Fig 2). Only 2 removals were
justified by an apparent cure of the stenosis. This was
a lower rate than that in the series of Martinez-Ballarin et al,10 in which 21 of 48 prosthesis removals
were justified by an apparent cure. Only 3 prostheses
had to be removed on day 0, and 1 on day 1, for bad
tolerance or ineffectiveness, and in the literature, the
rate of effectiveness of the stents on dyspnea is very
high: between 80% and 97%.4,11-13 Stents also significantly improved the scores on pulmonary function tests.14-16 However, the effect of stents on survival has been studied very little. Lemaire et al3 and
Xu et al17 showed that survival increased if external
radiotherapy was performed after TBS placement, as
compared to the survival with TBS placement alone.
However, no studies comparing etiologic treatment
with versus without a TBS were found in the liter-
Second Prosthesis
1 no second prosthesis
7 expandable covered prostheses
1 expandable uncovered prosthesis
4 nonexpandable prostheses
1 no second prosthesis
1 expandable covered prosthesis
4 nonexpandable prostheses
2 no second prosthesis
3 expandable covered prostheses
4 nonexpandable prostheses
1 expandable covered prosthesis
1 no second prosthesis
2 nonexpandable prostheses
1 no second prosthesis
1 nonexpandable prosthesis
1 no second prosthesis
1 expandable covered prosthesis
2 nonexpandable prostheses
2 expandable covered prostheses
1 expandable uncovered prosthesis
1 nonexpandable prosthesis
1 no second prosthesis
1 expandable covered prosthesis
2 nonexpandable prostheses
1 nonexpandable prosthesis
1 no second prosthesis
1 nonexpandable prosthesis
1 expandable uncovered prosthesis
1 no second prosthesis
1 nonexpandable prosthesis
2 no second prosthesis
2 expandable covered prostheses
1 nonexpandable prosthesis
ature. Relief of dyspnea in terminal palliative care
was initially one of the major indications for use of
a TBS, because TBSs are particularly effective in increasing short-term survival and quality of life.18
The main disadvantage of the TBS is the high incidence of complications, which in published series
ranges from 13%3 to 72%.12 Dasgupta et al,19 in a
series of 37 patients, reported the incidence of complications in a more relevant way: the longer a patient has a TBS, the greater the risk of complications. Dasgupta et al19 thus calculated the incidence
of complications in terms of patient-months (probability of a complication in 1 patient in a month). We
also used this method in our study. Another term that
seems important to us is the incidence of “no complications.” Indeed, several complications often occur in 1 patient, and the incidence of complications
is therefore overestimated.
108
Buiret et al, Is Stent Brand Predictive Factor of Complications?
TABLE 4. ANALYSIS OF PREDICTIVE FACTORS
FOR STENT REMOVAL
Fig 2. Time of stent removal by cause of stenosis.
The principal complications are migrations, granuloma, sputum retention, in-stent tumoral growth or
overgrowth, and hemoptysis. Chhajed et al20 even
showed that patients with tracheobronchial stenosis
after pulmonary transplantation had reduced survival in the event of a TBS complication.
Every prosthesis has advantages and disadvantages (Table 5), and no ideal prosthesis exists. Our
experience in TBS use enables us to put forward a
number of reasons why certain stent brands have
greater rates of complications and removal than others.
Rush prostheses are very effective, and can be cut
to the required length for the trachea, as well as for
the bronchi. They do not move easily and are not
very prone to cause infection. They are not expandable and therefore are sometimes difficult to place.
Some cases of granuloma at their ends, especially
the lower end, occur. They are easy to remove. A
problem sometimes arises in terms of the contact
with the wall of the trachea, as the stent’s diameter
is greater in the distal part than in the proximal part
(contrary to the dimensions of the trachea).
Novatech Y prostheses also can be cut to length,
but once dilated, the force they exert is lower than
that of the Rush prosthesis. They are sometimes difficult to place (nonexpandable prostheses); they are
a little prone to cause infection, but are effective.
The patented studs do nothing to prevent migration
and hamper positioning after insertion. This stent is
easy to remove. Novatech Silmet prostheses are generally the best for the majority of tracheal stenoses,
but the release technique could be improved. They
are prone to cause infections, but are quite easy to
remove.
Cook prostheses are very rigid and bring effec-
Univariate
Analysis
Multivariate
Analysis
HR
HR
Female sex
0.688
Age
1
Malignancy
1.479
Causes
*
Type of prosthesis
Nonexpandable
1
prostheses
Expandable covered
0.591
prostheses
Expandable uncovered 0.546
prostheses
Prosthesis brand
Nonexpandable prostheses
Aboulker
13.152
Cook
2.573
Novatech Y
8.424
Rush Y
11.101
Expandable covered prostheses
Wallstent
1.472
Hanarostent
4.152
Novatech Silmet
7.062
Tracheobronxane
5.081
Ultraflex
1
Expandable uncovered prostheses
Wallstent
10.31
p
0.232
0.971
0.315
0.612
0.218
0.62
*
0.009
p
0.431
0.612
0.001
10.424
8.701
88.268
122.902
2.496
9.372
17.750
10.626
1
57.8
*Hazard ratios (HRs) are not specified, because they would crowd
Table, but parameter is not significant.
tive dilation and good calibration. They allow excellent drainage of secretions, and granulomas are rare.
Release requires experience, because they need to
be pushed forward gently. Removal is feasible but
difficult. The problem is the very limited range of
diameters and lengths, which reduces indications.
Two prostheses should not be placed end-to-end (because of the occurrence of stenosis between them)
or one inside the other (to avoid any migration of
the unit).
Hanarostent prostheses are no longer used, as they
do not provide enough expansion. Wallstent and Ultraflex prostheses, covered or not, are rather easy to
place, but cause severe mucosal reactions at their extremities, with granuloma and infections. Tracheobronxane prostheses are not very practical and do
not expand sufficiently, and the patented studs have
no utility; they should never be placed between the
cartilages of the trachea, and contact between the
prosthesis and the tracheal wall must be avoided, as
there is a risk of sputum accumulation at the lower
end.
Buiret et al, Is Stent Brand Predictive Factor of Complications?
109
TABLE 5. ADVANTAGES AND DISADVANTAGES OF TYPES OF TRACHEOBRONCHIAL STENTS
Advantages
Disadvantages
Expandable uncovered stents
Immediate relief of dyspnea
Little sputum retention
Effectiveness on intrinsic and extrinsic
obstructions
Ease of deployment
Low migration rate
Possible in fibroscopy in outpatients
Low thickness, good internal/external diameter
ratio
No interference with mucociliary clearance
Improvement of quality of life
Growth within stent or at ends
Not usable with tracheoesophageal fistula
Difficulty of removal after epithelialization
Infection
Hemorrhage
Granuloma
Fracture or crushing if external force is too great
Diminution of length if increase of diameter
Y stent not available
Stent fatigue and fracture
Expandable covered stents
Treatment of extrinsic compression
Treatment of tracheoesophageal fistula
Less migration than nonexpandable prostheses
Facility and precision of placement under video
control
Large choice of diameters
Low thickness, good internal/external diameter
ratio
Good biocompatibility
Less risk of tumoral ingrowth (except at ends)
compared to expandable uncovered stents
Easier to remove
Less interference with mucociliary clearance
than nonexpandable stents
Less fracture than expandable uncovered stents
Granulation at ends
Migration
Obstruction by sputum
Repositioning and extraction more difficult, but
remain possible
Expensive
Can occlude small bronchi
Diminution of length if increase of diameter
Exactitude of placement with fluoroscopy is
clearly less than with visual installation
Radial force could cause necrosis and formation
of tracheoesophageal fistula
High rate of minor complications (bad
positioning, granulation, retention of secretions)
Fracture or crushing if external force is too great
Only Y stent available
Nonexpandable stents
Facility of placement and repositioning
Facility of removal
Resistance to external compression
Y stents for perfect adaptation to carina
Minimal granulation
Resistance to tumor ingrowth or intrastent
granulation
Can allow hemostasis in event of perendoscopic
hemorrhage
Not expensive
Migration
Thickness of wall, reduction of internal diameter
Retention or adherence of sputum
General anesthesia and rigid bronchoscopy
Rigid
Interference with mucociliary clearance
Less effective than expandable stents in case of
compression mass because of lack of expansion
No conformation to sinuosity of tracheobronchial
tree
Three techniques for TBS placement are currently used. Initially described with Dumon nonexpandable prostheses, placement during bronchoscopy under general anesthesia is the most frequently
used technique. It ensures relatively safe access to
the bronchial tree, allows better endoscopic visualization of the tumor, and gives more room to the
operators.2 In our opinion, suspension laryngoscopy under general anesthesia provides a wider range
of possibilities than does bronchoscopy21,22 (laser,
electrocoagulation, dilation). Moreover, the instruments can be changed more quickly than with bronchoscopy,22 because both hands are free.
Moreover, laryngoscopy makes it easier to determine the location of the stenosis compared to the
carina, and to measure stenosis length and diameter
compared with the trachea diameter. The disadvantages include the risk of dental avulsion injury, the
risk of oropharyngeal injury,22 and the need for general anesthesia. More recently, the practice of prosthesis placement under local anesthesia, possibly
without fluoroscopy,23 has spread. This technique
seems sure, fast, and well tolerated, and is usable
in emergencies24 and in outpatients, thus decreasing the length of hospitalization and costs.25 The
main disadvantages include frequent malpositioning13,24,26 leading to immediate repositioning, frequent fluoroscopy,2 limited interventional possibilities due to the low number of working channels,22
and an inability to use a laser correctly.27 We have
no experience of this technique in our center.
110
Buiret et al, Is Stent Brand Predictive Factor of Complications?
Acknowledgment: The authors thank Philip Bastable, English Medical Department, University of Burgundy, Dijon, France, for the
English translation.
REFERENCES
1. Montgomery WW. T-tube tracheal stent. Arch Otolaryngol 1965;82:320-1.
2. Makris D, Marquette CH. Tracheobronchial stenting and
central airway replacement. Curr Opin Pulm Med 2007;13:27883.
3. Lemaire A, Burfeind WR, Toloza E, et al. Outcomes of
tracheobronchial stents in patients with malignant airway disease. Ann Thorac Surg 2005;80:434-8.
4. Walser EM, Robinson B, Raza SA, Ozkan OS, Ustuner
E, Zwischenberger J. Clinical outcomes with airway stents for
proximal versus distal malignant tracheobronchial obstructions.
J Vasc Interv Radiol 2004;15:471-7.
5. Bolliger CT, Sutedja TG, Strausz J, Freitag L. Therapeutic bronchoscopy with immediate effect: laser, electrocautery, argon plasma coagulation and stents. Eur Respir J 2006;27:125871.
6. Chin CS, Litle V, Yun J, Weiser T, Swanson SJ. Airway
stents. Ann Thorac Surg 2008;85:S792-S796.
7. Walser EM. Stent placement for tracheobronchial disease. Eur J Radiol 2005;55:321-30.
8. Gaissert HA, Grillo HC, Wright CD, Donahue DM, Wain
JC, Mathisen DJ. Complication of benign tracheobronchial
strictures by self-expanding metal stents. J Thorac Cardiovasc
Surg 2003;126:744-7.
9. Saad CP, Murthy S, Krizmanich G, Mehta AC. Selfexpandable metallic airway stents and flexible bronchoscopy:
long-term outcomes analysis. Chest 2003;124:1993-9.
10. Martinez-Ballarin JI, Diaz-Jimenez JP, Castro MJ, Moya
JA. Silicone stents in the management of benign tracheobronchial stenoses. Tolerance and early results in 63 patients. Chest
1996;109:626-9.
11. Ryu YJ, Kim H, Yu CM, Choi JC, Kwon YS, Kwon OJ.
Use of silicone stents for the management of post-tuberculosis
tracheobronchial stenosis. Eur Respir J 2006;28:1029-35.
12. Kapoor BS, May B, Panu N, Kowalik K, Hunter DW.
Endobronchial stent placement for the management of airway
complications after lung transplantation. J Vasc Interv Radiol
2007;18:629-32.
13. Monnier P, Mudry A, Stanzel F, et al. The use of the covered Wallstent for the palliative treatment of inoperable tracheobronchial cancers. A prospective, multicenter study. Chest
1996;110:1161-8.
14. Miyazawa T, Yamakido M, Ikeda S, et al. Implantation of
ultraflex nitinol stents in malignant tracheobronchial stenoses.
Chest 2000;118:959-65.
15. Vergnon JM, Costes F, Bayon MC, Emonot A. Efficacy
of tracheal and bronchial stent placement on respiratory functional tests. Chest 1995;107:741-6.
16. Abdullah V, Yim AP, Wormald PJ, van Hasselt CA. Dumon silicone stents in obstructive tracheobronchial lesions: the
Hong Kong experience. Otolaryngol Head Neck Surg 1998;118:
256-60.
17. Xu X, Tajima H, Ishioh M, et al. Study on the treatment
of tracheobronchial stenosis using expandable metallic stents. J
Nippon Med Sch 2001;68:318-27.
18. Vonk-Noordegraaf A, Postmus PE, Sutedja TG. Tracheobronchial stenting in the terminal care of cancer patients with
central airways obstruction. Chest 2001;120:1811-4.
19. Dasgupta A, Dolmatch BL, Abi-Saleh WJ, Mathur PN,
Mehta AC. Self-expandable metallic airway stent insertion employing flexible bronchoscopy: preliminary results. Chest 1998;
114:106-9.
20. Chhajed PN, Malouf MA, Tamm M, Spratt P, Glanville
AR. Interventional bronchoscopy for the management of airway
complications following lung transplantation. Chest 2001;120:
1894-9.
21. Merrot O, Buiret G, Gleizal A, Poupart M, Pignat JC.
Management of tracheobronchial stenoses with endoprostheses: experience with 103 patients and 11 models. Laryngoscope
2008;118:403-7.
22. Eckel HE, Berendes S, Damm M, Klussmann JP, Wassermann K. Suspension laryngoscopy for endotracheal stenting.
Laryngoscope 2003;113:11-5.
23. Herth F, Becker HD, LoCicero J III, Thurer R, Ernst A.
Successful bronchoscopic placement of tracheobronchial stents
without fluoroscopy. Chest 2001;119:1910-2.
24. Spinelli P, Cerrai FG, Spinelli A. Palliation of tracheobronchial malignant obstruction with metal stents in emergency
conditions. Minerva Chir 1998;53:373-6.
25. Yerushalmi R, Fenig E, Shitrit D, et al. Endobronchial
stent for malignant airway obstructions. Isr Med Assoc J 2006;
8:615-7.
26. Shin JH, Kim SW, Shim TS, et al. Malignant tracheobronchial strictures: palliation with covered retrievable expandable nitinol stent. J Vasc Interv Radiol 2003;14:1525-34.
27. Vergnon JM. Supportive care. Endoscopic treatments
for lung cancer [in French]. Rev Mal Respir 2008;25:3S1603S166.
Posterior Glottic Diastasis:
Mechanically Deceptive and Often Overlooked
Steven M. Zeitels, MD; Alessandro de Alarcon, MD; James A. Burns, MD;
Gerardo Lopez-Guerra, MD; Robert E. Hillman, PhD
Dysphonia secondary to posterior glottic aerodynamic incompetence can often be recognizable acoustically, but difficult to document visually. This mechanical impairment in posterior glottic closure is the result of injury caused by airway instrumentation. The difficulty of recognition of this entity is due to posterior supraglottic soft tissue that obscures
the complete view during posterior glottic adduction, the lack of a structural organization of the cricoarytenoid region
injury that leads to this disorder, and the lack of nomenclature. A retrospective assessment was done on 3 patients who
underwent surgical reconstruction to correct posterior phonatory incompetence subsequent to laryngotracheal intubation.
All 3 had sustained an injury to the cricoarytenoid joints, and 2 of the 3 had undergone paraglottic space medialization
laryngoplasty that failed to solve the posterior glottic insufficiency. New procedures were designed and performed in
these patients to correct the posterior glottic incompetence and are described: laryngofissure and partial posterior cricoid
resection, endoscopic pharyngoepiglottic-aryepiglottic fold advancement-rotation flap with interarytenoid interposition,
and interarytenoid submucosal implant augmentation. Although the academic literature is replete with reports describing
stenosis resulting from impaired cricoarytenoid joint abduction, the term glottic diastasis provides nomenclature for the
inability to normally adduct the arytenoid cartilages. The initial experience with surgical reconstruction is preliminary,
but encouraging.
Key Words: cricoid cartilage, dysphonia, hoarseness, intubation, laryngofissure, laryngoplasty, larynx, stenosis, vocal
cord, vocal fold.
been elegantly described by Vijayasekaran et al3 and
Benjamin and Holinger.4 To the best that we could
determine, Bastian and Richardson5 provided the
sole report to focus on the vocal deficits associated
with inadequate phonatory adduction subsequent to
endotracheal tube–induced posterior glottic injury.
Benjamin and Holinger4 provided an intraoperative
photograph illustrating the nature of the injury.
INTRODUCTION
Vocal dysfunction secondary to mechanical trauma of the posterior glottis from laryngotracheal reconstruction and/or airway stents is familiar to most
laryngeal surgeons. The inadequate posterior glottal
closure can be audibly perceived as breathy dysphonia. However, the diminished airway caliber appropriately supersedes vocal dysfunction as the focus
of treating clinicians. Indwelling stents have long
been used to reestablish an adequate laryngeal airway when posterior glottal injury has resulted in
stenosis.1,2
In our experience, permanent hoarseness subsequent to laryngotracheal intubation is substantially
less recognized and understood than is incompetent
posterior glottic closure caused by indwelling stents
used to treat stenosis, caused by laryngotracheal reconstruction with a posterior cricoid cartilage graft
interposition, or caused by cancer resection.6-9
Remarkably, there is a population of patients who
have permanent posterior glottic injury with a sustainable airway caliber but develop impaired cricoarytenoid adduction as a result of limited laryngotracheal intubation. The complex mechanisms
of injury in the posterior glottis and cricoarytenoid
joints resulting from laryngotracheal intubation have
In patients who have had laryngotracheal intubation, the dysphonia secondary to posterior glottic
aerodynamic incompetence can often be recognizable acoustically, but is difficult to document visu-
From the Department of Surgery, Harvard Medical School (Zeitels, Burns, Lopez-Guerra, Hillman), and the Center for Laryngeal Surgery and Voice Rehabilitation, Massachusetts General Hospital (Zeitels, Burns, Lopez-Guerra, Hillman), Boston, Massachusetts, and
the Department of Otolaryngology–Head and Neck Surgery, University of Cincinnati College of Medicine and Cincinnati Children’s
Hospital Medical Center, Cincinnati, Ohio (de Alarcon). This work was supported in part by the Eugene B. Casey Foundation and the
Institute of Laryngology and Voice Restoration.
Presented at the meeting of the American Broncho-Esophagological Association, Las Vegas, Nevada, April 28-29, 2010.
Correspondence: Steven M. Zeitels, MD, Center for Laryngeal Surgery and Voice Rehabilitation, Massachusetts General Hospital,
One Bowdoin Square, 11th Floor, Boston, MA 02114.
111
112
Zeitels et al, Posterior Glottic Diastasis
ally.5 Our observations revealed that patients often
sound substantially more breathy than they appear
on laryngoscopic examination. This is because the
upper arytenoid and corniculate regions tilt forward
in an attempt to close the glottic incompetence, and
they obscure visualization of a posterior keyhole aperture at the vocal fold level. Identifying the submucosal posterior glottic injury can be further opacified when the patient has also sustained vocal fold
denervation.
Despite the commonly used term glottic stenosis
for inadequate abductory opening of the glottic aperture, we could not identify an ideal term for inadequate adductory closure of the posterior glottis.
Considering this scenario, we consulted with an individual skilled in the Greek language, and on the
basis of that discussion and further analysis, determined that an optimal term for inadequate interarytenoid closure/apposition would be posterior glottic diastasis. Diastasis can be defined as a
permanent separation of structures that should be
joined, which describes arytenoid closure during
phonatory glottic adduction. Koufman et al10 utilized the term arytenoid hypertelorism for the malpositioned arytenoid cartilage associated with a
chondrosarcoma. This was accurate for tumor-related arytenoid displacement, which is analogous
to displacement of the midface and orbit, but is not
ideal for a scenario in which the cricoarytenoid joints
are not displaced. In our experience, patients who
present with dysphonia secondary to glottic diastasis
have all sustained injury from mechanical instrumentation of the posterior glottic region. Although we
have commonly observed idiopathic glottic stenosis,
we have not identified idiopathic glottic diastasis.
Limited recognition of posterior glottic diastasis
arises from the frequent difficulty in visually identifying laryngotracheal intubation–induced posterior glottic incompetence, along with the lack of diagnostic terminology. Even when clinicians clearly
recognize the harsh, breathy dysphonia, two typical strategies are utilized to treat the patient. Most
commonly, the patients are deemed to have “hyperfunctional” voices, and voice therapy is administered. Not uncommonly, those who have sustained
posterior glottic injury from an endotracheal tube
have underlying reflux laryngitis,11 so this chronic
mucosal problem is treated as well. Alternatively,
but not exclusively, the presumed diminished glottic closure is deemed to be neurogenic paresis, so
that a medialization of the anterior paraglottic phonatory mucosal region is performed. The arytenoid
cartilage frequently retains substantial mobility, and
arytenoid repositioning procedures are not done, so
as not to diminish the airway caliber for the promise
of some enhanced vocal function.
Although we had recognized this entity with increasing frequency, we had difficulty designing the
optimal procedure that would rectify the problem
without placing the cricoarytenoid abductory function at risk in one or both vocal folds. Conceptually, we concluded that there were three possible reconstructive possibilities, and an example of each
of these is described herein. The documentation
detailed herein should provide the readership with
clarity about this entity and lead to development of
further-enhanced reconstructive options.
MATERIALS AND METHODS
An Institutional Review Board–approved retrospective assessment was done on 3 patients who
underwent surgical reconstruction to correct posterior phonatory incompetence. The procedures were
classified as follows: 1) laryngofissure and partial
posterior cricoid resection; 2) endoscopic pharyngoepiglottic-aryepiglottic fold advancement-rotation flap with interarytenoid interposition; and 3)
interarytenoid submucosal implant augmentation.
All 3 patients had sustained an intubation-induced
injury to the cricoarytenoid joints. Two of the 3 had
undergone paraglottic space medialization laryngoplasty that failed to solve the posterior glottic insufficiency.
CASE REPORTS
Patient 1. This patient (Figs 1 and 2) was a 35year-old woman who developed an acute myasthenic crisis and was orotracheally intubated for 3 days
in the intensive care unit. After that intubation, her
voice never recovered. She had severe breathy dysphonia. Her initial treating laryngologist noted the
posterior glottic incompetence and diagnosed it as
bilateral paresis. She underwent paraglottic space
augmentations initially, with office-based Cymetra
(Micronized AlloDerm) injections, followed by bilateral silicone medialization. The patient reported
that the transcervical medialization resulted in worse
vocal quality. On presentation to us, there was normal and brisk symmetric abduction of both arytenoid
cartilages, and there was bilateral limited adduction
that did not allow for interarytenoid closure.
When she presented, the expanded paraglottic
space from the left implant was easily seen through
the ventricular surface of the vocal fold mucosa.
There was substantial loss of interarytenoid soft tissue, and the outlining of the cricoid cartilage was
easily seen posteriorly; the glottis was leaking air
posteriorly during phonatory adduction. In an attempt to close the keyhole aperture posteriorly, a
Zeitels et al, Posterior Glottic Diastasis
A
B
C
D
E
F
113
Fig 1. (Patient 1) A) Office telescopic image during full abduction. Note contour swelling of left vocal fold from submucosal implant in paraglottic space. B) Office telescopic examination during adduction. Note substantial glottic incompetence. C) Microlaryngoscopic examination of pharyngoepiglottic fold. Aryepiglottic fold flap is being sutured into
interarytenoid region. D) Early postoperative office telescopic examination shows fibrinous exudate in left aryepiglottic
fold region. Left vocal fold is seen distantly in central aspect of left side of image. E) Office telescopic examination approximately 3 weeks after operation during full abduction. F) Office telescopic examination approximately 3 weeks after
operation during full adduction.
long pharyngoepiglottic and aryepiglottic fold advanced rotation flap was made. The tip of that flap,
which started from the lateral oropharyngeal wall,
was sutured into the interarytenoid region.
This procedure was done without altering the
interarytenoid muscle or the capsule of either cricoarytenoid joint. In the immediate postoperative
period, the patient’s voice was dramatically better.
Flexible laryngoscopic examination revealed that
the posterior keyhole aperture had been closed by
the flap. On the first postoperative day, the voice had
deteriorated in part, and the patient reported having coughed extensively during the prior evening.
Laryngoscopy revealed that the distal tip of the flap
had dissociated from the interarytenoid region, presumably from the vigorous coughing and the newly
established aerodynamic competency. It was elected not to reposition the flap, because only one suture had become dissociated and the voice was still
114
Zeitels et al, Posterior Glottic Diastasis
A
Fig 2. (Patient 1) A) Laryngofissure has
been performed. Suction cannula is positioned between inferior posterior cricoid incision edges on perichondrium.
Hook is being used to withdraw inferior
aspect of strip of cricoid cartilage from
posterior cricoid plate. B) After removal of posterior cricoid strip, Prolene sutures are used to reapproximate edges of
posterior cartilage. C) Office telescopic
examination during abduction, approximately 3 weeks after operation. Glottis
is well healed. Remnant of anterior laryngofissure can be seen through glottic aperture. Interarytenoid region during full abduction is diminished by approximately 3 mm, which was thickness
of posterior cricoid cartilage strip. D)
Office telescopic examination on same
day as C, now showing substantial adductory improvement.
B
C
substantially improved over the preoperative state.
Furthermore, there was no pathway to preclude the
same event from occurring again.
After healing, there was substantial improvement
in the patient’s vocal quality, timbre, projection,
and stamina, but they were certainly not ideal. After utilizing her voice for a while, she deemed it to
be functional but not optimal and inquired whether
anything further could be done. Subsequently, a laryngofissure was performed after a precise, microlaryngoscopically controlled separation of the anterior commissure tendon. Three millimeters of the
posterior cricoid region was removed, and the cricoid cartilage was reapproximated posteriorly.
A tracheotomy was performed, and the tube was
left in place throughout the perioperative period
with the cuff inflated so as to avoid subcutaneous
emphysema. The cuff was deflated at 7 days, and
the tracheotomy tube was removed at 8 days. The
patient’s postoperative healing was uneventful, and
her voice was dramatically improved because of the
D
substantially improved closure of her interarytenoid
region. As was to be expected, she has 3 mm of diminished abductory airway caliber; however, it has
not caused significant airway symptoms, even during moderate athletic exercise.
Patient 2. This patient (Fig 3) was a 62-year-old
woman who developed encephalopathy and was
orotracheally intubated for 8 days. After a complete
neurologic recovery from that process, she was noted to have a severely breathy voice with diminished
mobility of both arytenoid cartilages and bilateral
granulomas. She underwent laryngeal electromyography, of which the findings were normal, and removal of the granulomas was done. Subsequently,
the patient was referred for management of bilateral cricoarytenoid joint fixation, which was approximately 7 months subsequent to the intubation.
Her clinic examination revealed that both arytenoid cartilages were in a fixed and abducted position
at approximately 7.0 mm. She had a very breathy
phonation quality and could partially “sphincter”
Zeitels et al, Posterior Glottic Diastasis
A
B
C
D
115
Fig 3. (Patient 2) A) Office flexible laryngoscopic examination at full abduction. B) Same examination as in A,
during attempts at adduction, shows that larynx is “sphinctering.” However, there is minimal alteration of interarytenoid region because of cricoarytenoid fixation. C) Office flexible laryngoscopic examination at approximately 1 month. As in patient 1, interarytenoid distance at full abduction is now diminished by approximately 3 mm.
D) Same examination as in C, during adduction, reveals that although patient cannot adduct arytenoid cartilages,
she can “sphincter” laryngeal musculature to achieve substantial closure of phonatory mucosa.
the anterior phonatory glottis so that there was limited phonatory mucosal vibration from high subglottic pressures. Because of the aerodynamic incompetence, she had a very weak voice. The patient generally had a sedentary lifestyle, even before the encephalopathy, and had limited aerodynamic exercise
requirements. The glottic diastasis was unequivocal,
given the normal electromyographic findings and
the history of intubation. A transoral-transcervical
laryngofissure with resection of 3 mm of posterior
cricoid cartilage was done on this patient as well.
Afterward, she maintained a 4.5-mm interarytenoid
airway that was sufficient for normal activities.
However, with sphincteric closure of the phonatory region, along with her extremely pliable vibratory mucosa, she had dramatic improvement in vocal
function. In the 6 months after the operation, she has
not had airway difficulties. She has not had symptoms of sleep apnea; however, on deep inspiration,
she has mild stridor.
Patient 3. The patient (Figs 4 and 5) was a 53-yearold woman who underwent lung transplantation and
had laryngotracheal intubation for approximately
10 days. She also sustained left vocal fold paralysis. The initial treating laryngeal surgeon performed
multiple Radiesse (calcium hydroxylapatite microspheres) injection medialization procedures in the
paraglottic space with the hope of closing the glottic
gap. An injection for excessive paraglottic space did
not solve the posterior glottic incompetence and actually compounded the dysphonia, because the normally pliable phonatory mucosa became extremely
stiff from the excessive expansion of the paraglottic
space.
This patient was treated in a 2-stage fashion. At
the initial procedure, a microlaryngoscopic exploration was done with a Hunsaker jet-ventilation catheter. A superior-surface cordotomy was done on the
left vocal fold. An excessive amount of Radiesse
was immediately encountered and extracted from
the paraglottic soft tissue.
The superior vocal fold incision was sutured
closed, and a lipoinjection12 was done to replace the
volume-depleted paraglottic region. Microlaryngo-
116
Zeitels et al, Posterior Glottic Diastasis
A
B
C
D
E
F
Fig 4. (Patient 3) A) Office telescopic examination during full abduction demonstrates paralyzed left vocal fold. B) Office telescopic examination during adduction reveals poor glottic closure and submucosal swelling of left vocal fold from
prior implant of paraglottic space. C) After substantial anesthesia of corniculate region, flexible laryngoscopic examination of interarytenoid region shows significant loss of interarytenoid soft tissue caused by previous use of endotracheal
tube, revealing outline of cricoid cartilage. D) Flexible laryngoscopic examination in close proximity to left vocal fold
reveals submucosal Radiesse (calcium hydroxylapatite microspheres) bulging below mucosa of superior surface of vocal
fold. E) Microlaryngoscopic examination shows dissection of Radiesse from paraglottic space through superior-surface
epithelial cordotomy. F) Large piece of Radiesse is being removed through epithelial cordectomy.
scopic examination of the interarytenoid region revealed that the endotracheal tube had caused substantial loss of soft tissue. Therefore, a mucosal incision was made at the cephalad end of the cricoid
cartilage, and a cold-instrument pocket was created. Then, a portion of the Radiesse that had been
extracted from the left paraglottic space was reimplanted in the interarytenoid region to diminish the
posterior glottic keyhole aperture while not injuring
the cricoarytenoid joint capsules.
Despite some vocal improvement, there was persistent posterior glottic incompetence due to the malpositioned arytenoid cartilage, as well as the incompletely corrected extensive loss of interarytenoid
soft tissue from the previous indwelling endotracheal tube. At a second procedure, an adduction
arytenopexy13,14 was done with monitored intrave-
Zeitels et al, Posterior Glottic Diastasis
A
B
C
D
117
Fig 5. (Patient 3) A) After removal of Radiesse, epithelial cordotomy is sutured closed. B) Microlaryngoscopic examination of interarytenoid region demonstrates submucosal pocket that has been made just inside left interarytenoid region,
with piece of Radiesse in place. Mucosal incision is being sutured closed. C) Office telescopic examination during abduction, after second-stage procedure in which adduction arytenopexy was performed. D) Office telescopic examination
demonstrates substantially enhanced adductory closure.
nous sedation. This diminished the posterior glottic incompetence that was secondary to the abducted
arytenoid cartilage in the paralyzed vocal fold. After
the operation, the patient had further enhancement
of her vocal function.
Voice Assessment. Standard voice assessments
were performed before and after surgery in 2 of the
3 patients. The third patient did not return for postsurgical voice assessment because of medical complications related to transplantation treatment. The
assessments included videostroboscopy, completion of the Voice-Related Quality-of-Life Survey
(V-RQL),15 and objective acoustic and aerodynamic
measures of vocal function. Because of their reliance
on subjective judgments, observations from videostroboscopic recording were not used as formal data
and are only included in the context of discussing
the results of vocal function measures. The details
of the protocol for obtaining acoustic and aerodynamic measures of vocal function have previously
been described.13 For the 2 patients with complete
voice assessment data, postsurgical measures were
compared descriptively with presurgical measures
and with historical norms.13
RESULTS
Despite the disparate mechanical injuries, all 3
patients reported substantial improvement in their
vocal function. None have had a complication or
have required airway intervention as a result of the
narrowed glottic aperture. The presurgical and postsurgical voice assessment results for the 2 patients
with complete data are summarized in the Table.
We utilized a subset of 4 primary voice assessment
measures to evaluate the impact of the surgery on
vocal function. The measures included the overall
V-RQL score, the average fundamental frequency
(vocal pitch) during reading of a standard passage,
the ratio between noise and harmonic energy during
sustained vowels (noise-to-harmonics ratio [NHR];
voice quality), and the average oral airflow rate
(in liters per second) during comfortable sustained
vowel production as an estimate of average glottic
airflow.
Patient 1 displayed dramatic postsurgical improvements in V-RQL score, NHR, and airflow; the
V-RQL score increased by 70 points, and the NHR
and airflow rate decreased from highly abnormal
values to within normal limits. The fundamental fre-
118
Zeitels et al, Posterior Glottic Diastasis
PRESURGICAL AND POSTSURGICAL VOICE ASSESSMENT RESULTS FOR TWO PATIENTS WITH COMPLETE DATA
V-RQL Score
Patient
Preop
Postop
1
2
Norms
2
28
72
40
100 = best
F0 (Hz)
Preop
Postop
183
178
Aphonic
192
180-230
NHR
Preop
Airflow (L/s)
Postop
0.37
0.17
Aphonic
0.21
≤0.19
Preop
Postop
0.792
0.147
Aphonic
0.530
0.05-0.25
V-RQL — Voice-Related Quality-of-Life Survey; F0 — fundamental frequency; NHR — noise-to-harmonics ratio.
quency for patient 1 was in the low-borderline-normal range before and after surgery. Patient 2 was actually aphonic before surgery, so it was not possible
to obtain acoustic and aerodynamic measures. After
surgery, patient 2 did display measurable phonation,
with an improvement of 12 points in V-RQL score,
a normal fundamental frequency, and abnormally
high values for NHR and airflow rate.
DISCUSSION
Glottic stenosis, with its associated airway narrowing and breathing restriction due to diminished
arytenoid abduction, has been recognized since the
origin of our specialty with widespread adoption of
mirror laryngoscopy.16 This was not uncommonly
due to severe infectious upper respiratory tract diseases that afflicted the laryngeal airway. As necessary, tracheotomy17 was performed to bypass the
obstructed laryngeal airway. In the late 19th century, O’Dwyer18 perfected the placement of tubular indwelling stents. Although the size, shape, and
constituent material of these stents have changed
and developed over the past 150 years, their efficacy in reestablishing airway patency is well established. O’Dwyer’s work provided a foundation for
20th-century laryngotracheal intubation.19
Laryngotracheal intubation can result in posterior glottic phonatory incompetence during arytenoid
adduction, visually observed as a keyhole aperture
(Fig 4C), because of a combination of one or more
of three factors. These include loss of arytenoid cartilage3,5 from pressure necrosis, injury of the medial cricoarytenoid joint,3,5 and loss of normal interarytenoid mucosal soft tissue. Typically, an indwelling endotracheal tube or stent leads to pressure
necrosis of the posterior glottic tissues, resulting in
the aforementioned findings. A classic posterior cricoid split with cartilage interposition is actually designed to create posterior glottic diastasis to remedy
the more morbid and common problem of stenotic
airway obstruction.
Although a submucosal loss of arytenoid cartilage
and submucosal fibrosis of the medial cricoarytenoid
joint are difficult to observe, loss of interarytenoid
redundant mucosa that is normally present can be
observed with careful office-based laryngoscopy
(Fig 4C). This mucosa, which often becomes hyperplastic in patients with reflux,20 stretches thin during interarytenoid abduction and corrugates to close
the extreme posterior glottis during adduction. Flexible laryngoscopy with substantial topical anesthesia for extreme posterior positioning of the distal
scope shaft is often necessary to observe the posterior glottis keyhole-aperture incompetence, because
the supraglottic arytenoid region and associated corniculate mucosa often tilt forward and obscure the
site of glottic air leakage.
In 2 of the 3 cases described herein, unsuccessful
prior attempts at glottic closure were made by augmenting the paraglottic space. Patient 1 had multiple bilateral Cymetra injections followed by bilateral silicone medialization procedures. These procedures had a detrimental vocal effect, because they
did not close the posterior glottic gap, yet they increased the surface tension and stiffness of the phonatory mucosa. A similar mechanical outcome occurred in patient 3, who underwent multiple injections of Radiesse into the paraglottic region.
Reconstructive Options. Although there has been
limited recognition of the vocal deficits characterized herein as posterior glottic diastasis, a phonosurgical solution has not been forwarded, in part because of the unavoidable concern that reconstructive efforts to enhance phonation would negatively
affect the airway and possibly lead to intubation or
tracheotomy.
Although we had only preliminary success in this
small pilot group of patients, we present this report
because the clinical concepts are robust. The patient
population who would benefit from repair is small
and geographically widespread, so establishing optimal reconstructive techniques will require multiinstitutional incremental advancements that integrate different surgeons’ creative innovations.
There are two feasible approaches for closing
posterior glottic phonatory incompetence while preserving an adequate posterior glottic airway. One
would be to replace the lost soft tissue in the interarytenoid region at the cephalad edge of the posterior cricoid cartilage, and the second would be to
diminish the inter–cricoid facet distance and there-
Zeitels et al, Posterior Glottic Diastasis
by bring the arytenoid cartilages closer together. We
approached the problem initially by augmenting the
lost soft tissue and had limited success. Thus far, it
has been more effective to remove a strip of cricoid
cartilage posteriorly. This latter procedure can be
understood as the inverse of a posterior cricoid split
and distraction with a rib graft.6
We replaced lost soft tissue in the interarytenoid
region endoscopically in 2 of the 3 cases. In the patient who underwent the pharyngoepiglottic and
aryepiglottic fold flap, the voice was initially much
stronger; however, a coughing episode with the
newly competent posterior glottic valve partially
dislodged the distal flap from the interarytenoid region. Her voice maintained some improvement over
her preoperative state, but became substantially better after a second procedure with removal of a strip
of posterior cricoid cartilage.
The patient who underwent placement of Radiesse
in a submucosal interarytenoid pocket also gained
improvement; however, there was not enough redundant mucosa in that region after the intubation
injury to substantially diminish the aperture of the
aerodynamically incompetent posterior glottis. Because of her underlying vocal fold paralysis, she
also required repositioning of her arytenoid body on
the cricoid facet to optimize her voice.
Thus far, our impression is that if the airway caliber is deemed to be adequate for a patient’s needs,
dysphonia from glottic diastasis is best treated by
a laryngofissure and removal of a vertical strip of
posterior cricoid cartilage to diminish the inter–
cricoid facet distance. Not surprisingly, this is the
inverse of the procedure undergone by those who
have a posterior cricoid cartilage interposition6,7
to splay the posterior cricoid facets to treat glottic
and subglottic stenosis. In patient 2, the endotracheal tube functioned as a stent, completely fixing the
cricoarytenoid joints in a widely abducted position,
rather than leading to the more common scenario of
stenosis. In our view, she could tolerate some nar-
119
rowing of her airway aperture, because she was relatively sedentary. With removal of a posterior 3-mm
strip of cricoid cartilage, she was able to maintain
a comfortable airway yet had remarkable improvement in her voice through general sphincteric closure of her phonatory glottic valve.
Complete voice assessment data were available
for 2 of the 3 patients. In both cases, there was clear
improvement in postsurgical vocal function. One of
the patients went from being aphonic to having measurable phonation that was abnormally breathy (increased airflow rate and NHR) and was associated
with a surprisingly small improvement in the subject’s perception of her vocal status (V-RQL). The
other patient displayed dramatic improvements in
both objective (NHR and airflow rate) and self-rated
(V-RQL) measures of vocal function. Taken together, the results from these preliminary cases provide
evidence that the new surgical procedures described
here have had a decidedly positive impact on vocal
function.
CONCLUSIONS
Limited awareness of the physiology underlying
breathy dysphonia associated with vocal fold adductory tissue injuries from laryngotracheal intubation
has been compounded by difficulty in visually identifying the often-subtle submucosal trauma. This is
in contrast to the glottic aerodynamic incompetence
that is easily seen and heard after surgical treatment
of glottic cancer, stenosis, or bilateral paralysis. The
academic literature is replete with reports describing
stenosis resulting from impaired cricoarytenoid joint
abduction, yet we were unable to find a term for impaired posterior glottic adductory closure. We think
that posterior glottic diastasis can serve as nomenclature for this disorder, and the initial experience
with surgical reconstruction is encouraging. Several
treatment strategies have been demonstrated herein
that should catalyze further multi-institutional innovations and efforts.
REFERENCES
1. Mackenzie M. Dilators of the larynx. In: Diseases of the
pharynx, larynx, and trachea. New York, NY: William Wood,
1880:192-5.
5. Bastian RW, Richardson BE. Postintubation phonatory
insufficiency: an elusive diagnosis. Otolaryngol Head Neck
Surg 2001;124:625-33.
2. Jackson C, Jackson CL. Dilation with elastic rubber core
molds. In: The larynx and its diseases. Philadelphia, Pa: WB
Saunders, 1937:447-54.
6. Cotton RT. The problem of pediatric laryngotracheal
stenosis: a clinical and experimental study on the efficacy of
autogenous cartilaginous grafts placed between the vertically
divided halves of the posterior lamina of the cricoid cartilage.
Laryngoscope 1991;101(suppl 56):1-34.
3. Vijayasekaran S, Sances R, Cotton R, Elluru RG. Changes in the cricoarytenoid joint induced by intubation in neonates.
Arch Otolaryngol Head Neck Surg 2006;132:1342-5.
4. Benjamin B, Holinger LD. Laryngeal complications
of endotracheal intubation. Ann Otol Rhinol Laryngol Suppl
2008;117(suppl 200):1-20.
7. Rutter MJ, Cotton RT. The use of posterior cricoid grafting in managing isolated posterior glottic stenosis in children.
Arch Otolaryngol Head Neck Surg 2004;130:737-9.
8. Smith ME, Marsh JH, Cotton RT, Myer CM III. Voice
120
Zeitels et al, Posterior Glottic Diastasis
problems after pediatric laryngotracheal reconstruction: videolaryngostroboscopic, acoustic, and perceptual assessment. Int J
Pediatr Otorhinolaryngol 1993;25:173-81.
9. Zeitels SM, Hillman RE, Franco RA, Bunting GW. Voice
and treatment outcome from phonosurgical management of early
glottic cancer. Ann Otol Rhinol Laryngol Suppl 2002;111(suppl
190):1-20.
10. Koufman JA, Cohen JT, Gupta S, Postma GN. Cricoid
chondrosarcoma presenting as arytenoid hypertelorism. Laryngoscope 2004;114:1529-32.
11. Koufman JA. The otolaryngologic manifestations of gastroesophageal reflux disease (GERD): a clinical investigation
of 225 patients using ambulatory 24-hour pH monitoring and an
experimental investigation of the role of acid and pepsin in the
development of laryngeal injury. Laryngoscope 1991;101(suppl
53):1-78.
12. Burns JA, Kobler JB, Zeitels SM. Microstereo-laryngoscopic lipoinjection: practical considerations. Laryngoscope
2004;114:1864-7.
13. Zeitels SM, Hochman I, Hillman RE. Adduction arytenopexy: a new procedure for paralytic dysphonia with implications for implant medialization. Ann Otol Rhinol Laryngol
1998;107(suppl 173):1-24.
14. Zeitels SM, Mauri M, Dailey SH. Adduction arytenopexy
for vocal fold paralysis: indications and technique. J Laryngol
Otol 2004;118:508-16.
15. Hogikyan ND, Wodchis WP, Terrell JE, Bradford CR,
Esclamado RM. Voice-related quality of life (V-RQOL) following type 1 thyroplasty for unilateral vocal fold paralysis. J Voice
2000;14:378-86.
16. Solis-Cohen J. Diseases of the throat: a guide to diagnosis and treatment. New York, NY: William Wood, 1880:58693.
17. Louis A. Louis on bronchotomy. Presented before the
Royal Academy of Surgery of France in 1784. In: Ottley D, ed.
Observations on surgical diseases of the head and neck. London: The Sydenham Society, 1848:214-78.
18. O’Dwyer J. Chronic stenosis of the larynx treated by a
new method. Med Rec (NY) 1886;29:641.
19. Zeitels SM, Broadhurst MS, Akst LM, Lopez-Guerra
G. The history of tracheotomy and intubation. In: Myers EN,
Johnson JT, eds. Tracheotomy: airway management, communication, and swallowing. San Diego, Calif: Plural Publishing,
2007:1-21.
20. Jackson C, Jackson CL. Pachydermia of the larynx. In:
The larynx and its diseases. Philadelphia, Pa: WB Saunders,
1937:194-8.
Neurologic Variant Laryngomalacia Associated With Chiari
Malformation and Cervicomedullary Compression: Case Reports
Rajanya S. Petersson, MD; Nicholas M. Wetjen, MD; Dana M. Thompson, MD, MS
Two infants presented with intermittent stridor and evidence of laryngomalacia on flexible laryngoscopy. The first was a
10-month-old girl who had undergone 3 supraglottoplasty surgeries at an outside institution, without long-term resolution
of symptoms. She was found during our evaluation to have a Chiari malformation. Laryngomalacia symptoms resolved
after suboccipital decompression and C1 laminectomy, and the patient remained symptom-free at 6-month follow-up.
The second infant was a 24-day-old boy with velocardiofacial syndrome who was found to have posterior cervicomedullary junction compression at the level of C1. He underwent C1 laminectomy for decompression of the brain stem, which
resulted in immediate resolution of symptoms, and he remained symptom-free at 12-month follow-up. Neurologic abnormalities have been reported in up to 50% of infants with laryngomalacia. As such, brain stem dysfunction should be
considered among the causes of laryngomalacia during evaluation, especially in patients with failure of supraglottoplasty.
Both of these infants had resolution of symptoms after their neurosurgical procedures.
Key Words: brain stem compression, Chiari malformation, laryngomalacia, stridor.
INTRODUCTION
toms resolved after decompressive surgery.
Laryngomalacia is a condition of weak laryngeal
tone resulting in dynamic prolapse of supraglottic
tissue into the airway. This causes inspiratory stridor
and airway obstruction. The diagnosis is suspected
in the presence of symptoms of inspiratory stridor
that worsens with agitation, supine positioning,
feeding, and crying, and is confirmed by flexible
laryngoscopy. Laryngoscopic findings may include
prolapse into the airway of supra-arytenoid mucosa or the arytenoid cartilages themselves, foreshortened aryepiglottic folds, and/or an omega-shaped or
retroflexed epiglottis. In up to 20% of infants with
laryngomalacia, there have been associated neurologic abnormalities.1
Laryngeal stridor has been reported in association with Chiari II malformations (caudal displacement of the brain stem into the cervical spinal canal
associated with myelomeningocele), with the stridor most often caused by vocal fold paralysis.2-9 In
some of these cases, the actual laryngeal abnormality was not discovered.3,4 We present 2 cases of infants with neurologic variant laryngomalacia. One
infant was found to have an underlying Chiari I malformation, and the other was found to have cervicomedullary compression by a C1 posterior arch. In
both patients, laryngomalacia and associated symp-
CASE REPORTS
Case 1. A 10-month-old girl with intermittent
inspiratory stridor, episodes of cyanosis, and aspiration throughout infancy presented to our clinic.
The stridor worsened with supine positioning. She
had undergone supraglottoplasty 3 times at an outside institution, with only temporary improvement
in symptoms. After the first supraglottoplasty, the
episodes of cyanosis resolved. She had temporary
relief of other symptoms, but shortly after surgery
she experienced worsening stridor and aspiration.
This prompted a second supraglottoplasty 5 months
later, from which she had relief of symptoms for
only 2 days. Overall, the stridor was improved after
her first surgery, but was still present during sleep
and with agitation. A third supraglottoplasty was
performed 1 month later, again with only 2 days of
symptom relief. The patient presented to us 7 days
after her last surgery.
At presentation, she had inspiratory stridor during sleep without desaturations. The stridor was
high-pitched during sleep, and the patient’s breathing sounded raspy during the daytime while she was
active. She was not experiencing cyanosis or retractions. She was still aspirating thin liquids, and was
From the Departments of Otorhinolaryngology (Petersson, Thompson) and Neurologic Surgery (Wetjen), Mayo Clinic, and the Division of Pediatric Otorhinolaryngology, Mayo Clinic and Mayo Eugenio Litta Children’s Hospital (Thompson), Rochester, Minnesota.
Presented at the meeting of the American Broncho-Esophagological Association, Las Vegas, Nevada, April 28-29, 2010.
Correspondence: Dana M. Thompson, MD, Division of Pediatric Otorhinolaryngology, Mayo Clinic, 200 First St SW, Rochester,
MN, 55905.
121
122
Petersson et al, Neurologic Variant Laryngomalacia
A
B
Fig 1. (Case 1) Magnetic resonance imaging. A) Preoperative scan shows Chiari I malformation with descent of
cerebellar tonsils below foramen magnum. B) Scan performed 6 months after neurosurgical decompression shows
resolution.
on a thickened formula diet. She was on twice-daily proton pump inhibitor and nighttime histamine 2
receptor antagonist therapy. Flexible laryngoscopy
revealed a slightly omega-shaped epiglottis without significant supra-arytenoid tissue prolapse; the
lowest oxygen saturation was 90% during the examination. Significant cobblestoning of the posterior pharyngeal wall was noted. A functional endoscopic evaluation of swallowing study revealed laryngeal penetration of thickened formula with resultant cough, and occasional bolus passage without
penetration or aspiration; there was direct laryngeal
penetration and aspiration with thin liquid.
As it is unusual for an infant to continue to have
symptoms of laryngomalacia after a third supraglottoplasty, a thorough evaluation was planned.
Computed tomography (CT) scans of the neck and
chest showed no anatomic anomalies and no signs
of chronic aspiration. Magnetic resonance imaging
(MRI) of the head was scheduled to look for any
intracranial or skull base disorders as contributing
factors and did, in fact, reveal a Chiari I malformation, with descent of the cerebellar tonsils below the
level of the foramen magnum with significant compression within the foramen magnum (Fig 1A). The
patient was referred to the pediatric neurosurgery
department at this time, and further plans for a pH
study and overnight polysomnography were put on
hold.
She was taken to the operating room in conjunction with our pediatric neurosurgery colleagues for
microlaryngoscopy and rigid bronchoscopy and for
suboccipital decompression and C1 laminectomy.
Her airway examination revealed an omega-shaped
epiglottis with the lateral surfaces folding into the
airway and producing stridor and foreshortened
aryepiglottic folds, but no arytenoid prolapse. Rigid
bronchoscopy revealed mucosal edematous changes but a widely patent airway. On postoperative day
(POD) 1, the patient still had her baseline level of
inspiratory stridor, with some improvement on POD
2, when she was discharged from the hospital. By
her 1-week follow-up appointment, her stridor had
resolved. She was still taking thickened formula, but
without any symptoms of aspiration. Six months after her decompressive surgery, she was completely
asymptomatic; she had no stridor or signs of aspiration with thin liquids, was tolerating table foods,
was in the 95th percentile for weight, and was no
longer requiring proton pump inhibitors or histamine 2 receptor antagonists. Her postoperative MRI
no longer showed descent of her cerebellar tonsils
(Fig 1B).
Case 2. A 24-day-old boy with velocardiofacial
syndrome presented with intermittent biphasic stridor, nasopharyngeal regurgitation, spitting up, and
brief cyanotic episodes that occurred with feeding.
Flexible laryngoscopy confirmed laryngomalacia
with supra-arytenoid tissue prolapse into the glottal inlet, foreshortened aryepiglottic folds, and a
slightly omega-shaped epiglottis (Fig 2A); he experienced desaturations into the mid-80% range during the examination. As such, overnight oximetry
was performed and revealed a mean oxygen saturation of 95% with a high of 100% but a low of 72%;
Petersson et al, Neurologic Variant Laryngomalacia
A
123
B
Fig 2. (Case 2) Flexible laryngoscopy. A) Preoperative laryngoscopy shows supra-arytenoid tissue prolapse obscuring
visualization of vocal folds, foreshortened aryepiglottic folds, and omega-shaped epiglottis on inspiration. B) One day
after neurosurgical decompression, significantly decreased prolapse of supra-arytenoid tissue allows visualization of vocal folds on inspiration.
an oxygen saturation of less than 90% was recorded
for 3.5% of the test duration. High-kilovoltage airway films and fluoroscopy were performed because
of the biphasic nature of the stridor, and the findings
were normal.
During a medical genetics evaluation, MRI revealed posterior cervicomedullary compression and
kinking of the brain stem (Fig 3), prompting evalu-
Fig 3. (Case 2) Preoperative magnetic resonance imaging demonstrates posterior cervicomedullary compression and kinking of brain stem.
ation by the pediatric neurosurgery department. A
CT scan performed to evaluate the bony anatomy
revealed incomplete ossification of the anterior arch
of C1, kinking of the upper cervical spinal cord by
the C1 posterior arch, and lateral displacement of
the C1 lateral masses with bony settling of the occiput onto C2. Dynamic MRI was then performed
to assess the stability of his craniovertebral junction; it demonstrated clear compression of the spinal
cord in neutral position and reduced compression
in extension. There was no translational instability
on flexion-extension plain radiographs or dynamic
MRI, and no anterior compression of the brain stem.
Decompression with C1 laminectomy followed by
temporary cervicothoracic external orthosis stabilization until occipitocervical stability could be demonstrated was recommended by the pediatric neurosurgery department.
The patient was taken to the operating room in
conjunction with our pediatric neurosurgery colleagues for microlaryngoscopy and rigid bronchoscopy and for C1 laminectomy. The airway examination revealed a slightly omega-shaped epiglottis,
foreshortened aryepiglottic folds, and significant
supra-arytenoid prolapse into the airway; the findings on rigid bronchoscopy were unremarkable. On
POD 1, his stridor at baseline had nearly resolved,
feeding had improved, and flexible laryngoscopy
showed significantly decreased prolapse of supraarytenoid tissues (Fig 2B), and the patient was discharged from the hospital. At his 2-week follow-up
visit, he was free of stridor and was no longer spit-
124
Petersson et al, Neurologic Variant Laryngomalacia
ting up with feeding. Flexible laryngoscopy revealed
a normally shaped and placed epiglottis and only
mild supra-arytenoid prolapse; there was no audible
stridor, even with agitation. Supraglottoplasty was
never required. The child remained symptom-free
at the 18-month follow-up, and cervical flexion-extension radiographs demonstrated no occipital C1-2
instability.
DISCUSSION
Laryngeal stridor due to vocal fold paralysis associated with Chiari II malformations (also known
as Arnold-Chiari malformations) has been commonly reported.2-9 There have been few reported
cases of laryngomalacia associated with Chiari malformations, and no reported cases of laryngomalacia caused by C1 posterior arch compression of the
cervicomedullary junction. Portier et al6 described
a case of an infant with a Chiari I malformation
(elongation of cerebellar tonsils through the foramen magnum) and laryngomalacia causing respiratory obstruction that resolved 10 days after posterior
fossa decompression. Their study found that major
abnormal laryngeal dynamics, most often vocal fold
paralysis, were found in 11% of infants with Chiari
II malformations and less than 1% of those with
Chiari I malformations. Neither of our patients had
vocal fold paralysis in addition to laryngomalacia.
Neurologic disease of any type is a well-known
comorbidity associated with laryngomalacia, with
reported rates of 8% to 50%, and is known to contribute to supraglottoplasty failure.1,10-17 Supraglottoplasty in general carries a high success rate, and
has been reported to approach 94%, even when revision is required.1,18 Thus, the fact that our first
patient failed to have complete resolution of symptoms after 3 supraglottoplasty procedures led us to
believe that she may have had an underlying neurologic disorder. It is important to consider potential
comorbidities and additional testing in infants with
failed supraglottoplasty, as it is an operation with
high success rates in the absence of comorbidities.
Recognizing that our second patient had MRI evidence of cervicomedullary compression that could
be decompressed by a neurosurgical procedure obviated the need for supraglottoplasty. This patient
might have undergone supraglottoplasty first, with
potential failure, if MRI had not been performed for
evaluation of anomalies associated with velocardiofacial syndrome.
The pathophysiology of brain stem dysfunction
associated with Chiari malformations has been attributed to either hydrocephalus or brain stem compression itself.6 Our patient with the Chiari I malformation did not have hydrocephalus, so we assume
that the dysfunction was related to physical compression. Knowledge of the neurologic pathways responsible for laryngeal function and tone is important in understanding how brain stem compression
may contribute to development of laryngomalacia.
There is a vagal nerve–mediated reflex called the
laryngeal adductor reflex,1 which is activated when
mechanoreceptors and chemoreceptors of the superior laryngeal nerve are stimulated, sending sensory
afferent information to the brain stem nuclei responsible for regulating respiration and swallowing. Integration between the nucleus tractus solitarius and
the nucleus ambiguus occurs in the brain stem. Efferent signals are sent via the vagus nerve to the larynx, causing vocal fold adduction and a swallow.
These efferent signals are also responsible for laryngeal tone.1 Thus, brain stem compression may affect efferent vagal responses, leading to decreased
laryngeal tone and laryngomalacia. Both of our patients had complete resolution of symptoms associated with laryngomalacia and no evidence of laryngomalacia on flexible laryngoscopy after their brain
stem decompressive procedures. This outcome may
be further evidence supporting the neurologic theory of laryngomalacia causation.
CONCLUSIONS
Neurologic abnormalities have been reported in
up to 50% of infants with laryngomalacia. As such,
brain stem dysfunction should be considered among
the causes of laryngomalacia prompting further
evaluation, especially in patients who have failed
supraglottoplasty or other underlying comorbidities
(eg, genetic syndrome, congenital anomalies). Both
of these infants had resolution of laryngomalaciaassociated symptoms after their brain stem decompression procedures.
REFERENCES
1. Thompson DM. Abnormal sensorimotor integrative function of the larynx in congenital laryngomalacia: a new theory of
etiology. Laryngoscope 2007;117(suppl 114):1-33.
2. Smith ME. The association of laryngeal stridor with meningo-myelocele. J Laryngol Otol 1959;73:188-90.
3. Fitzsimmons JS. Laryngeal stridor and respiratory obstruction associated with meningomyelocele. Arch Dis Child
1965;40:687-8.
4. Krieger AJ, Detwiler JS, Trooskin SZ. Respiratory function in infants with Arnold-Chiari malformation. Laryngoscope
1976;86:718-23.
5. Cochrane DD, Adderley R, White CP, Norman M, Steinbok P. Apnea in patients with myelomeningocele. Pediatr Neurosurg 1990-1991;16:232-9.
Petersson et al, Neurologic Variant Laryngomalacia
6. Portier F, Marianowski R, Morisseau-Durand MP, Zerah
M, Manac’h Y. Respiratory obstruction as a sign of brainstem
dysfunction in infants with Chiari malformations. Int J Pediatr
Otorhinolaryngol 2001;57:195-202.
7. Morley AR. Laryngeal stridor, Arnold-Chiari malformation and medullary haemorrhages. Dev Med Child Neurol 1969;
11:471-4.
8. Papasozomenos S, Roessmann U. Respiratory distress
and Arnold-Chiari malformation. Neurology 1981;31:97-100.
9. Holinger PC, Holinger LD, Reichert TJ, Holinger PH.
Respiratory obstruction and apnea in infants with bilateral abductor vocal cord paralysis, meningomyelocele, hydrocephalus,
and Arnold-Chiari malformation. J Pediatr 1978;92:368-73.
10. Hoff SR, Schroeder JW Jr, Rastatter JC, Holinger LD.
Supraglottoplasty outcomes in relation to age and comorbid
conditions. Int J Pediatr Otorhinolaryngol 2010;74:245-9.
11. Reddy DK, Matt BH. Unilateral vs bilateral supraglottoplasty for severe laryngomalacia in children. Arch Otolaryngol
Head Neck Surg 2001;127:694-9.
125
12. Olney DR, Greinwald JH Jr, Smith RJ, Bauman NM. Laryngomalacia and its treatment. Laryngoscope 1999;109:17705.
13. Toynton SC, Saunders MW, Bailey CM. Aryepiglottoplasty for laryngomalacia: 100 consecutive cases. J Laryngol
Otol 2001;115:35-8.
14. Denoyelle F, Mondain M, Gresillon N, Roger G, Chaudre
F, Garabedian EN. Failures and complications of supraglottoplasty in children. Arch Otolaryngol Head Neck Surg 2003;129:
1077-80.
15. Roger G, Denoyelle F, Triglia JM, Garabedian EN. Severe laryngomalacia: surgical indications and results in 115 patients. Laryngoscope 1995;105:1111-7.
16. McClurg FL, Evans DA. Laser laryngoplasty for laryngomalacia. Laryngoscope 1994;104:247-52.
17. Prescott CA. The current status of corrective surgery for
laryngomalacia. Am J Otolaryngol 1991;12:230-5.
18. Richter GT, Thompson DM. The surgical management of
laryngomalacia. Otolaryngol Clin North Am 2008;41:837-64,
vii.
Systematic Review of Endoscopic Airway Findings in
Children With Gastroesophageal Reflux Disease
Jason Glenn May, MD; Priyanka Shah, MA; Lori Lemonnier, MD; Gaurav Bhatti, MS;
Jovana Koscica, DO; James M. Coticchia, MD
Objectives: We performed a systematic review of published literature correlating findings on endoscopic evaluation of
the larynx and trachea in the pediatric population with the incidence of gastroesophageal reflux disease.
Methods: Eight articles were identified through a structured PubMed search of English-language literature using the
key terms laryngopharyngeal reflux, extraesophageal reflux, and gastroesophageal reflux. A systematic review was performed relating the presence of reflux in the pediatric population to findings on endoscopic airway evaluation. A covariant analysis was performed, and each study was weighted according to the number of available samples in that study as a
fraction of the total. Overall odds ratios and confidence intervals were computed for each endoscopic finding on the basis
of the documented absence or presence of gastroesophageal reflux disease.
Results: A correlation was seen between the endoscopic findings and the presence of reflux.
Conclusions: Arytenoid, postglottic, and vocal fold edema and erythema, lingual tonsil hypertrophy, laryngomalacia,
and subglottic stenosis are among the endoscopic findings most frequently identified in patients with gastroesophageal
reflux disease. Certain findings commonly encountered on endoscopic evaluation of the larynx and trachea in children
who present with respiratory symptoms do indeed demonstrate a correlation with the presence of laryngopharyngeal reflux disease and may indicate the need for antireflux therapy.
Key Words: extra-esophageal reflux, gastroesophageal reflux disease, laryngopharyngeal reflux.
INTRODUCTION
ration pneumonia, exacerbation of asthma, apnea,
stridor, croup, and laryngitis or hoarseness.2 A relationship has also been shown between GERD and
chronic lung diseases such as cystic fibrosis and
bronchopulmonary dysplasia.3 It has been postulated that more than 40% of children with GERD have
associated respiratory manifestations.4 The anatomic proximity of the larynx and supraglottis to refluxing material from the esophagus makes associated
airway findings a distinct possibility.
Reflux disease has become one of the most prevalent disease entities in the United States, with an
estimated 75 million people suffering from gastroesophageal reflux disease (GERD). In the pediatric
population, the prevalence has been reported to be as
high as 22%.1 Gastroesophageal reflux is defined as
a retrograde movement of gastric contents, including pepsin, bile, and pancreatic enzymes, into the
esophagus. Reflux beyond the esophagus is termed
laryngopharyngeal reflux (LPR). Extraesophageal
reflux disease refers to nongastrointestinal manifestations of reflux disease. There is increasing evidence that GERD, and more specifically LPR, is associated with other medical problems ranging from
chronic otitis media with effusion to airway abnormalities and esophageal dysfunction. A thorough
understanding of the pathophysiology involved and
its possible implications has become a necessity.
The classic symptoms for GERD include heartburn, regurgitation, and dyspepsia, whereas the presentation of LPR may be less predictable. In the pediatric population, airway symptoms include aspi-
The most significant difference between LPR and
GERD is heartburn, as the majority of children with
LPR do not have esophagitis or its primary symptom, heartburn.2 Common LPR symptoms include
frequent throat clearing or cough, globus or foreign
body sensation, and hoarseness, among others. Endoscopic findings that are commonly associated with
chronic inflammation include edema and erythema
of the arytenoids, redundancy of interarytenoid mucosa or pachydermia, and inflammatory changes of
the vocal folds. In contrast, GERD is associated with
heartburn and can occasionally be associated with
distal esophageal changes and metaplasia, such as
From the Division of Pediatric Otolaryngology, Department of Otolaryngology, Wayne State University, Detroit, Michigan.
Presented at the meeting of the American Broncho-Esophagological Association, Las Vegas, Nevada, April 28-29, 2010.
Correspondence: James M. Coticchia, MD, 4201 St Antoine, Suite 5E, Detroit, MI 48201.
126
May et al, Review of Gastroesophageal Reflux Disease
in Barrett’s esophagus. One key point in understanding these two disease entities is that both GERD and
LPR can occur in synchrony.
The possible connection between GERD and airway abnormalities remains a controversial topic.
Recently, there has been increasing evidence supporting a link between GERD or LPR and airway
disease. The difficulty in dealing with this topic is
in finding a cause-and-effect relationship between
GERD and airway disease. How does the clinician
tell whether GERD and airway disease merely coexist? Furthermore, some studies in the literature show
coexistence and possible connection, but few show
convincing causality. Several retrospective and prospective studies have demonstrated airway findings
characteristic of GERD- or LPR-positive pediatric
patients. These findings included arytenoid, interarytenoid, and postglottic edema and erythema, laryngomalacia, subglottic stenosis, cobblestoning of
the tracheal mucosa, and blunting of the carina. Yellon et al3 showed a correlation between several endoscopic airway findings and GERD, which included laryngomalacia, subglottic stenosis, and posterior
glottal edema and erythema. Their study also reported a correlation between GERD and airway symptoms such as cough, stridor, asthma, and croup.
Halstead5 reported a 35% reduction in the need
for airway surgery for subglottic stenosis if patients
received aggressive therapy for GERD. This study is
important in that it points to a causality of GERD in
airway disease. Aggressive treatment of a patient’s
reflux resulted in decreased airway disease and thus
a reduction in the need for airway surgery. Several
other studies have also pointed to treating reflux as
a means of improving the pediatric airway. Tashjian
et al6 investigated the effects of Nissen fundoplication on reactive airway disease. They showed that
effective management of GERD or LPR via Nissen
fundoplication decreased airway disease in children.
Common presenting symptoms for these children
included cough, dyspnea, and airway congestion.
The study showed that 75% of children who underwent Nissen fundoplication were off all reflux medications and, more importantly, were symptom-free.
Another study, by Suskind et al,7 showed the resolution of reflux-related otolaryngology symptoms after antireflux surgery. Otolaryngology symptoms included subglottic edema, subglottic stenosis, reflex
apnea, and recurrent croup. These studies showed
resolution of airway symptoms after surgical management of medically refractory reflux that pointed
to a cause-and-effect relationship between GERD
and airway disease. Unfortunately, no studies to date
have attempted to analyze and evaluate the current
literature correlating common airway findings on
127
endoscopy to the presence of GERD or LPR. The
purpose of this study was to perform a meta-analysis to evaluate the correlation of endoscopic findings
of LPR and GERD in the pediatric population. Our
hypothesis holds that there is an increasing body of
literature that supports a link between GERD and
endoscopic findings consistent with airway disease.
METHODS
A systematic review was performed of articles relating the presence of reflux disease in the pediatric
population to findings on endoscopic airway evaluation. These were identified through a structured
PubMed search of the English-language literature
using the key terms laryngopharyngeal reflux, extraesophageal reflux, and gastroesophageal reflux,
as well as subglottic stenosis and laryngomalacia.
Additional articles were obtained by hand-searching
the references listed in those papers identified by
the above search criteria. The literature search was
performed in late 2009 and represented the studies
available at that time. We included all articles found
by the above-mentioned literature search and hand
search method. We excluded articles studying strictly endoscopic findings in the pediatric population.
The result was a total of 20 articles.3-5,7-23 Each was
reviewed to determine the level of evidence, subject
population, methods of evaluation, results, and conclusions.
Fourteen of the 20 articles were used in the study.
Six could not be included in the statistical analysis because of an inability to compare them to the
other articles in the study. (Two articles were based
on different therapies administered to patients with
symptoms. Three articles did not include the numbers of patients with GERD for each airway finding reported, and instead only discussed percentages
of patients with GERD and certain airway findings.
One article looked only at patients who had a diagnosis of GERD and whose airway findings were
investigated.) Further, 6 additional articles were excluded because of numbers too small for comparison and unclear methodology. The result was 8 articles that were reviewed and submitted to meta-analysis.3-5,9,11,12,18,23
There are several ways in which clinicians measure GERD. The gold standard has shifted over the
years and is currently a 24-hour pH probe study.
Methods have included the following: evaluation
of esophageal biopsies on a histopathologic examination suggestive of GERD, barium reflux into the
esophagus on a barium swallow test, double and single pH probes showing a pH of less than 4 for 5% to
8% of time, and number of reflux episodes. Several
studies used multiple methods.
128
May et al, Review of Gastroesophageal Reflux Disease
Fig 1. Meta-analysis of arytenoid edema and erythema
and gastroesophageal reflux disease.4,11 CI — confidence interval. Asterisk — p < 0.05.
Fig 2. Meta-analysis of posterior glottal edema and erythema and gastroesophageal reflux disease.4,11 Asterisk
— p < 0.05.
The age ranges of the children used in the different studies varied tremendously. Some studies had
a wide range, evaluating children in ages ranging
from 10 days to 17 years. Other studies focused on a
very narrow age range.
sis revealed a correlation between 6 endoscopic airway findings and the presence of GERD. The airway findings that correlated to GERD in a statistically significant manner are listed below. Arytenoid
edema and erythema were found to correlate with
GERD with a relative risk of 2.46 (Fig 14,11). Lingual tonsil hypertrophy was found to correlate with
GERD with a relative risk of 2.24. Posterior glottal
edema and erythema were found to strongly correlate with GERD, with a relative risk of 3.19 (Fig
24,11). Subglottic stenosis was found to correlate
with the presence of GERD in 4 studies with a relative risk of 2.5 (Fig 34,9,11,16). Tracheal edema was
found to correlate with GERD with a relative risk
of 1.86. Vocal fold edema and nodules was found to
have the strongest correlation to GERD, with a relative risk of 12.15 (Fig 411,12). Epiglottic and supraglottic collapse were found to correlate with GERD,
but not in a statistically significant manner.
Moderated risk ratio analysis was performed on
4 variables as mentioned above. The variables that
were studied in at least 3 articles were included in
Several airway findings were observed in patients
from the studies. These included lingual tonsil swelling, postglottic edema and erythema, arytenoid edema and erythema, ventricle obliteration, true vocal
fold edema, laryngomalacia, tracheomalacia, vocal
fold lesions, subglottic stenosis, supraglottic stenosis, hypopharyngeal cobblestoning, narrowed trachea, posterior pharyngeal wall cobblestoning, innominate artery compression, choanal atresia, posterior glottal cleft, blunt carina, general edema and
erythema, and increased secretions. Some findings
were echoed in multiple reports. Five reports mentioned laryngomalacia, and 4 mentioned subglottic
stenosis.
Fisher’s exact test was used to evaluate the significance of contingency tables obtained from individual studies. The data from individual studies with
appropriate sample size were then combined, and
the significance was determined similarly. Weights
were assigned to a study based on its sample size
relative to the total sample size. Odds ratios with the
95% confidence interval along with the weights are
reported in the plots shown in Results.
Moderated risk ratios were calculated. This method examines risk factors (in this case, airway findings) evaluated in 4 or more studies. The closer the
value of the moderated risk ratio is to 1, the stronger
the correlation between the risk factor and GERD.
RESULTS
A meta-analysis and a moderated risk ratio analysis evaluated the correlation between endoscopic
larynx and airway findings and GERD. Meta-analy-
Fig 3. Meta-analysis of subglottic stenosis and gastroesophageal reflux disease.4,9,11,16 Asterisk — p < 0.05.
May et al, Review of Gastroesophageal Reflux Disease
Fig 4. Meta-analysis of vocal fold edema and nodules
and gastroesophageal reflux disease.11,12 Asterisk — p <
0.05.
the moderated risk ratio analysis. The moderated
risk ratio analysis illustrated the percentage chance
that a patient had GERD in the presence of specific
endoscopic airway findings. Patients with epiglottic
or supraglottic collapse had a 67% chance of GERD.
Patients with posterior glottal edema and erythema
had a 70% chance of having GERD. Patients with
subglottic stenosis had a 65% chance of having
GERD. Patients with vocal fold edema and/or nodules had an 88% chance of having GERD. Patients
with all 4 findings had a greater than 70% chance of
having GERD (Fig 53-5,9,11,12,18,23).
The reported incidence of GERD in patients presenting with respiratory symptoms within the 8
studies ranged from 31% to 86%. Laryngomalacia
and subglottic stenosis have been frequently examined in the literature in association with GERD, and
Fig 5. Moderated risk ratio comparing 4 variables that were compared in 3 or more of studies.3-5,9,11,12,18,23
129
were found concomitantly in 63% to 80% and 57%
to 95% of patients, respectively. Importantly, a correlation was identified by Giannoni et al18 between
the severity of laryngomalacia and that of GERD.
Additional endoscopic laryngeal findings frequently documented in the presence of GERD included
postglottic, arytenoid, and true vocal fold edema and
lingual tonsil hypertrophy. Furthermore, Carr’s triad11 of postglottic edema, loss of arytenoid contour,
and lingual tonsil hypertrophy was found in 61.5%
of patients with GERD by Mitzner and Brodsky,4
whereas Carr et al11 concluded that each finding
was pathognomonic for the disease. Last, treatment
options for GERD, including behavioral changes,
medical therapy, and surgical therapy, have proven
to have a significant impact on the improvement of
symptoms, as well as endoscopic findings, even resulting in the avoidance of airway surgery in 35% of
one study population.
DISCUSSION
Numerous investigators have performed endoscopic evaluations on children with a variety of clinical presentations that include recurrent croup, stridor, chronic cough, and even failure to thrive. Many
of these clinical studies have attempted to correlate
endoscopic findings with different diagnostic tests
for GERD, including vocal erythema and edema,
posterior glottal edema and erythema, arytenoid
edema, and even subglottic stenosis. The reflux of
gastric contents into the larynx and supraglottis (laryngopharynx) has been implicated as an inflamma-
130
May et al, Review of Gastroesophageal Reflux Disease
tory cofactor and a possible cause of many adult and
pediatric respiratory disorders.5 The gold standard
for diagnosing GERD is 24-hour pH probe monitoring. Endoscopic evaluation via direct laryngoscopy
and bronchoscopy is considered the gold standard
for diagnosing upper airway disease. Common findings associated with chronic inflammation include
supraglottic edema and erythema, vocal fold edema
and erythema, posterior glottal edema and erythema, tracheal edema and loss of tracheal rings, and,
finally, subglottic stenosis.
The studies reviewed in this report looked for endoscopic findings consistent with GERD and LPR.
Although there is no study that shows that specific
endoscopic findings are only caused by reflux disease, the literature has consistently investigated a
consistent set of airway findings. One could argue
that many of the same airway findings could be attributed to intubation trauma, but to do so would be
to ignore a large body of evidence that points to the
purported link between reflux and airway disease.
Both GERD and LPR cause chronic low-grade inflammation consistent with the pathogenesis of posterior glottal edema and erythema, vocal fold edema
and nodules, and even subglottic stenosis.
Halstead5 sought to show a correlation between
GERD and pediatric upper airway disorders. The
study looked at 51 children who underwent evaluation for GERD as a possible cofactor in their upper
airway disease. Forty-one of these children underwent 24-hour pH probe monitoring. Laryngoscopy
was performed to evaluate the children’s airway.
The study showed that all but 1 of the 41 children
probed had 10 or more episodes of pharyngeal reflux. It is believed that even 1 episode of reflux is
indicative of the presence of GERD. The laryngeal
appearance ranged from subtle edema to alarming
erythema and edema. Airway findings included true
vocal fold nodules, laryngomalacia, and subglottic
stenosis. The study showed that significant GERD
occurs in many pediatric upper airway disorders.
The study showed reductions in symptoms, in airway findings, and, most importantly, in the degree
of surgical intervention. The vast majority of children (90%) with subglottic stenosis showed a partial or complete resolution of GERD symptoms with
medical or surgical therapy. These children showed
a reduction in stridor, and 35% avoided airway surgery altogether. This study clearly shows a likely
cause-and-effect relationship between GERD and/
or LPR and airway disease.
Similar studies have shown a marked decrease
in airway disease and/or interventions by better controlling GERD and/or LPR. Tashjian et al6
performed a retrospective review of 24 children
who underwent Nissen fundoplication. Common
presenting symptoms of airway disease included
cough, dyspnea, airway congestion, and weight
loss, and the patients were taking multiple medications for reflux. Bronchoscopy showed lipid-laden
macrophages on bronchial washings. After the Nissen fundoplication, 75% of the patients stopped taking all medications and were symptom-free. Another study, by Suskind et al,7 showed the resolution of
reflux-related otolaryngological symptoms after antireflux surgery. Otolaryngological symptoms in the
study were defined as subglottic edema, subglottic
stenosis, reflex apnea, and recurrent croup. In all 14
patients, the otolaryngological symptoms resolved
after antireflux surgery. Again, these studies point to
a likely connection between GERD and airway disease. However, the study was limited by several factors. It was a retrospective study of patients who underwent evaluation for GERD. Also, there were no
further delineations of which specific airway findings, such as posterior glottal edema or erythema,
and vocal fold edema or erythema, correlated most
strongly with GERD.
Giannoni et al18 investigated the incidence of
GERD in patients with laryngomalacia. A prospective study was performed of 33 consecutive infants,
27 of whom were undergoing evaluation for GERD
via either esophagogram or 24-hour pH probe monitoring, with a new diagnosis of laryngomalacia.
GERD was observed in 64% of patients and was
associated with severe symptoms and a prolonged,
complicated course. Evaluation was done via questionnaire of the child’s caregiver. Endoscopic findings were broken down by severity of laryngomalacia found on laryngoscopy. The limitation of this
study is that it only examined children with a new
diagnosis of laryngomalacia. Also, once again, there
was no correlation done between specific endoscopic findings and the presence or severity of GERD.
Yellon et al3 investigated esophageal biopsy as
a means of diagnosing GERD. Also, the presence
of endoscopic findings such as posterior glottal erythema and edema, laryngomalacia, and subglottic
stenosis in children with GERD was investigated.
A historical cohort study was done on children who
underwent esophageal biopsy to investigate the presence of GERD. Esophageal biopsies were found to
be an effective method of diagnosing GERD. There
was also a correlation shown between GERD and
posterior glottal erythema and edema, laryngomalacia, and subglottic stenosis. A limitation of the study
is that it was a historical cohort study. Also, it only
investigated children who underwent esophageal bi-
May et al, Review of Gastroesophageal Reflux Disease
opsy.
Multiple methods were used to detect GERD in
the 8 studies that we reviewed. These methods included single pH probe monitoring, double pH
probe monitoring, and esophageal biopsy. However, it should be noted that 24-hour pH monitoring
is now considered the gold standard of detecting
GERD because of its 87% sensitivity, 93% specificity, and 92% accuracy. Chronic and uncontrolled
inflammation is now believed to play a key role in
the development of subglottic stenosis.
We sought to evaluate the studies currently in
the literature regarding the presence of GERD and
airway disease in children. Our systematic review
showed a statistically significant correlation between GERD or LPR and arytenoid edema and erythema, lingual tonsil hypertrophy, posterior glottal
edema and erythema, tracheal edema, vocal fold
edema and nodules, and, most importantly, subglottic stenosis. Certain endoscopic findings showed a
stronger correlation with GERD. Lingual tonsil hypertrophy and tracheal edema showed a statistically
significant correlation with GERD, but also showed
the weakest connection with GERD. Arytenoid edema and erythema and subglottic stenosis had a relative risk of around 2.5. In other words, children with
arytenoid edema and erythema were 2.5 times more
likely to have GERD than is the general population.
Interestingly, posterior glottal edema and erythema
showed a stronger correlation with GERD, with a
relative risk of greater than 3. Vocal fold edema and
nodules had the strongest correlation, with a relative
risk of greater than 12. The key point of this study
was elicited by the moderated risk ratio analysis of
all 8 studies. This analysis showed the likelihood of
having GERD with respective endoscopic findings.
Patients with epiglottic or supraglottic collapse had
a 67% chance of having GERD. Patients with posterior glottal edema and erythema had a 70% chance
of having GERD. Patients with subglottic stenosis
had a 65% chance of having GERD. Patients with
vocal fold edema and nodules had an 88% chance of
having GERD. Finally, patients with all 4 findings
had a greater than 70% chance of having GERD.
This study points to a prominent role of GERD in
the development and worsening of airway symptoms. If a patient is found to have vocal fold edema and nodules, then it is likely that GERD is present, as well. This is an important indicator as to how
that patient is to be managed in the future. Studies
have shown decreased morbidity of airway disease
with aggressive management of GERD.5 This is the
first study to show a statistically significant correlation between the presence of GERD and endoscopic
131
findings of arytenoid edema and erythema, lingual
tonsil hypertrophy, posterior glottal edema and erythema, tracheal edema, vocal fold edema and nodules, and, most importantly, subglottic stenosis
Any systematic review is limited, in that it is not
a randomized, controlled trial. Also, among the different studies, it is difficult to ensure consistency
in terminology among raters: what constitutes erythema, what constitutes edema, etc. Also, a metaanalysis is somewhat limited by its very design.
Comparing various studies and drawing conclusions from multiple studies as a group can be difficult at best, and impossible at worst. The studies
were done by multiple groups on different patient
populations and using different criteria. Finally, any
patient population that undergoes endoscopic evaluation is subject to bias, because those children had
airway symptoms severe enough to necessitate an
airway procedure.
Future studies should include a randomized, controlled trial investigating endoscopic findings and
their possible correlation with GERD. Also, few
studies have attempted to compare the effects of aggressive treatment of GERD and its impact on the
outcome and resolution of endoscopic airway findings.
CONCLUSIONS
This systematic review has shown endoscopic
airway findings to correlate with the presence of
GERD or LPR. It showed a correlation between
GERD or LPR and arytenoid edema and erythema,
lingual tonsil hypertrophy, posterior glottal edema
and erythema, tracheal edema, vocal fold edema and
nodules, and, most importantly, subglottic stenosis.
Also, a moderated risk ratio analysis showed that
children with arytenoid edema and erythema, posterior glottal edema and erythema, subglottic stenosis, and vocal fold edema and nodules had a more
than 70% chance of having GERD. Multiple studies
have shown that effectively treating GERD lessens,
if not resolves, airway disease and symptoms. Certain findings commonly encountered on endoscopic
evaluation of the larynx and trachea in children who
present with respiratory symptoms do indeed demonstrate a correlation with the presence of GERD,
and may indicate that antireflux therapy should be
considered in the treatment of this population of patients. Prior studies have shown that aggressive antireflux management reduces the amount of airway
surgery necessary for these patients.7 The correlation of endoscopic airway findings and GERD in
the 8 articles systematically reviewed by our study
points to the necessity of reflux management in children with the airway findings discussed.
132
May et al, Review of Gastroesophageal Reflux Disease
REFERENCES
1. Brodsky L, Carr MM. Extraesophageal reflux in children. Curr Opin Otolaryngol Head Neck Surg 2006;14:387-92.
diatric patients. Int J Pediatr Otorhinolaryngol 2005;69:110912.
2. Walner DL, Stern Y, Gerber ME, Rudolph C, Baldwin CY,
Cotton RT. Gastroesophageal reflux in patients with subglottic
stenosis. Arch Otolaryngol Head Neck Surg 1998;124:551-5.
3. Yellon RF, Coticchia J, Dixit S. Esophageal biopsy for
the diagnosis of gastroesophageal reflux–associated otolaryngologic problems in children. Am J Med 2000;108(suppl 4a):
131S-138S.
4. Mitzner R, Brodsky L. Multilevel esophageal biopsy in
children with airway manifestations of extraesophageal reflux
disease. Ann Otol Rhinol Laryngol 2007;116:571-5.
5. Halstead LA. Gastroesophageal reflux: a critical factor
in pediatric subglottic stenosis. Otolaryngol Head Neck Surg
1999;120:683-8.
6. Tashjian DB, Tirabassi MV, Moriarty KP, Salva PS. Laparoscopic Nissen fundoplication for reactive airway disease. J
Pediatr Surg 2002;37:1021-3.
7. Suskind DL, Zeringue GP III, Kluka EA, Udall J, Liu
DC. Gastroesophageal reflux and pediatric otolaryngologic
disease: the role of antireflux surgery. Arch Otolaryngol Head
Neck Surg 2001;127:511-4.
8. Bibi H, Khvolis E, Shoseyov D, et al. The prevalence
of gastroesophageal reflux in children with tracheomalacia and
laryngomalacia. Chest 2001;119:409-13.
9. Halstead LA. Role of gastroesophageal reflux in pediatric
upper airway disorders. Otolaryngol Head Neck Surg 1999;120:
208-14.
10. Little JP, Matthews BL, Glock MS, et al. Extraesophageal pediatric reflux: 24-hour double-probe pH monitoring of
222 children. Ann Otol Rhinol Laryngol Suppl 1997;106(suppl
169):1-16.
11. Carr MM, Nagy ML, Pizzuto MP, Poje CP, Brodsky LS.
Correlation of findings at direct laryngoscopy and bronchoscopy with gastroesophageal reflux disease in children: a prospective study. Arch Otolaryngol Head Neck Surg 2001;127:36974.
12. Carr MM, Abu-Shamma U, Brodsky LS. Predictive value of laryngeal pseudosulcus for gastroesophageal reflux in pe-
13. Matthews BL, Little JP, McGuirt WF Jr, Koufman JA.
Reflux in infants with laryngomalacia: results of 24-hour double-probe pH monitoring. Otolaryngol Head Neck Surg 1999;
120:860-4.
14. Walner DL, Holinger LD. Supraglottic stenosis in infants
and children. A preliminary report. Arch Otolaryngol Head
Neck Surg 1997;123:337-41.
15. Walner DL, Stern Y, Gerber ME, Rudolph C, Baldwin CY,
Cotton RT. Gastroesophageal reflux in patients with subglottic
stenosis. Arch Otolaryngol Head Neck Surg 1998;124:551-5.
16. Stroh BC, Faust RA, Rimell FL. Results of esophageal
biopsies performed during triple endoscopy in the pediatric patient. Arch Otolaryngol Head Neck Surg 1998;124:545-9.
17. Gumpert L, Kalach N, Dupont C, Contencin P. Hoarseness and gastroesophageal reflux in children. J Laryngol Otol
1998;112:49-54.
18. Giannoni C, Sulek M, Friedman EM, Duncan NO III.
Gastroesophageal reflux association with laryngomalacia: a
prospective study. Int J Pediatr Otorhinolaryngol 1998;43:1120.
19. Belmont JR, Grundfast K. Congenital laryngeal stridor
(laryngomalacia): etiologic factors and associated disorders.
Ann Otol Rhinol Laryngol 1984;93:430-7.
20. Mandell DL, Yellon RF. Synchronous airway lesions and
esophagitis in young patients undergoing adenoidectomy. Arch
Otolaryngol Head Neck Surg 2007;133:375-8.
21. Ungkanont K, Friedman EM, Sulek M. A retrospective
analysis of airway endoscopy in patients less than 1-month old.
Laryngoscope 1998;108:1724-8.
22. Block BB, Brodsky L. Hoarseness in children: the role of
laryngopharyngeal reflux. Int J Pediatr Otorhinolaryngol 2007;
71:1361-9.
23. Carr MM, Nguyen A, Poje C, Pizzuto M, Nagy M, Brodsky L. Correlation of findings on direct laryngoscopy and bronchoscopy with presence of extraesophageal reflux disease. Laryngoscope 2000;110:1560-2.
Correlation of Vocal Fold Nodule Size in Children and
Perceptual Assessment of Voice Quality
Roger C. Nuss, MD; Jessica Ward; Lin Huang, PhD; Mark Volk, DMD, MD;
Geralyn Harvey Woodnorth, MA
Objectives: We examined the relationship between the size of vocal fold nodules and perceptual rating of voice quality
in children.
Methods: We carried out an Institutional Review Board–approved retrospective study in a voice clinic within a tertiarycare pediatric medical center. We studied children seen between 2000 and 2009 with a primary diagnosis of vocal fold
nodules as the cause of their voice disturbance. Pediatric vocal fold nodule size was rated with a published validated
scale, and voice quality was rated on the Consensus Auditory-Perceptual Evaluation of Voice scale.
Results: One hundred forty-five patients met the inclusion criteria. Small nodules were noted in 23% of patients, medium
nodules in 39%, and large nodules in 37%. Univariate and multivariate analyses demonstrated a statistically significant
relationship (p < 0.05) between vocal fold nodule size and rated perceptual qualities of overall severity of voice disturbance, roughness, strain, pitch, and loudness. With the exception of loudness, as vocal fold nodule size increased, the
mean value of perceptual characteristics became larger. The age of the patient was a significant factor associated with the
overall severity of the voice disturbance and roughness.
Conclusions: The overall severity of a child’s voice disturbance and qualities of roughness, strain, pitch, and loudness
have a strong correlational relationship with pediatric vocal fold nodule size, which is suggestive of causality.
Key Words: CAPE-V scale, child, vocal fold nodule, voice quality.
In addition, it is well recognized that some children
improve with no intervention — only natural physical growth and maturity.
INTRODUCTION
Vocal fold nodules are widely appreciated to
be the most common and important physical finding associated with dysphonia in pediatric patients.
Approximately 40% of patients seen in a pediatric
tertiary care voice clinic are diagnosed with vocal
fold nodules1 (also Nuss et al, unpublished observations). There may be a number of underlying factors
creating an environment in which nodules are more
likely to develop. Such contributing causes include
laryngopharyngeal reflux, vocal hyperfunction, vocal overuse and abuse, and even a genetic predisposition toward the development of nodules.2-4
One area of difficulty has been in understanding
whether the size of a vocal fold nodule in a child actually matters, and whether our treatment attempts
should even consider the size of nodules as an important factor. It is possible to objectively rate the
size of vocal fold nodules in children by use of a
validated scale.7 An experienced clinician’s perceptual assessment of a child’s voice quality can also
be objectively reported with a validated scale.8 The
goal of this study was to determine whether there
is any relationship between vocal fold nodule size
in children and perceptual assessment of their voice
quality.
The management of pediatric vocal nodules has
not been standardized, but is generally based on the
severity of the voice disorder. Many of the treatment options for children have been inferred from
adult therapies, including voice rest, voice therapy,
treatment of laryngopharyngeal reflux, and medical
management of any associated condition that may
interfere with the resolution of vocal fold nodules.5,6
METHODS
The Committee on Clinical Investigation at Children’s Hospital Boston approved this retrospective
study. The Voice Clinic at Children’s Hospital Boston maintains a database of children evaluated since
From the Department of Otolaryngology and Communication Enhancement (Nuss, Ward, Volk, Woodnorth) and the Clinical Research
Program (Huang), Children’s Hospital Boston, and the Department of Otology and Laryngology, Harvard Medical School (Nuss, Ward,
Volk, Woodnorth), Boston, Massachusetts.
Correspondence: Roger C. Nuss, MD, Dept of Otolaryngology and Communication Enhancement, Children’s Hospital Boston, 300
Longwood Ave, Boston, MA 02115.
133
134
Nuss et al, Vocal Nodule Size & Voice Quality
Fig 1. Distribution of patient population by age and nodule size.
Fig 2. Mean values of perceptual characteristics according to nodule size.
1996. The inclusion criteria for this study included
ages 2 through 18 years, examinations recorded between 2000 and 2009, a recorded high-quality digital videolaryngoscopic examination confirming the
presence of vocal fold nodules, and documented
perceptual ratings of voice quality on the Consensus
Auditory-Perceptual Evaluation of Voice (CAPE-V)
scale.8
95% consistency, ie, a high degree of intrarater reliability.
Perceptual assessment of voice quality was performed by an experienced pediatric voice and speech
pathologist using the CAPE-V scale.8 The characteristics measured included roughness, strain, pitch,
loudness, breathiness, and overall severity.
Transnasal videostroboscopic examination was
performed in all patients. An FNL-10RP3 fiberoptic nasolaryngoscope (KayPENTAX, Lincoln Park,
New Jersey) was used for transnasal examinations in
children 13 years of age and above; a KayPENTAX
FNL-7RP3 fiberoptic nasolaryngoscope was used
in children 2 to 12 years of age. Topical anesthesia
was achieved with the application of a 1:1 mixture
of 0.05% oxymetazoline hydrochloride solution and
4% lidocaine hydrochloride sprayed into the nasal
passage. Videostroboscopy examinations were recorded with a KayPENTAX Digital Video Stroboscopy System (Model 9295).
A 1-way analysis of variance (ANOVA) was used
to analyze the relationship of nodule size and perceptual voice variables (roughness, strain, pitch,
loudness, breathiness and overall severity) at the
univariate level. A t-test was used to see whether
nodule type (sessile or discrete) was significantly
related to perceptual voice measures. Age groups
(2.0 to 5.9 years, 6.0 to 11.9 years, and more than
12 years of age) and gender were also tested at the
univariate level by use of an ANOVA and a t-test
to determine whether they were significantly associated with perceptual voice variables. Nodule size,
nodule contour, age, and gender were all included
for multivariate analysis using factorial ANOVA.
Main effects and interaction terms were checked for
statistical significance, then least square means for
perceptual voice variables were computed adjusted
for the significant factors.
Patients were separated into three different age
groups associated with the levels of development:
2.0 to 5.9 years, 6.0 to 11.9 years, and more than
12 years of age. The senior author (R.C.N.) rated
vocal fold nodule size as small, moderate, or large,
according to a previously validated scale.7 Additionally, nodule contours were rated as being either sessile or discrete by use of this scale. Nodule sizes and
contours were later re-rated by the senior author,
blinded to the original rating. There was a more than
All of our tests were 2-sided and set 0.05 as a
standard type I error. The statistical software package SAS 9.2 was used for analysis (SAS Institute,
Cary, North Carolina).
TABLE 1. MEAN SCORE OF PERCEPTUAL CHARACTERISTICS, BASED ON CAPE-V SCALE, BY NODULE SIZE
Nodule Size
Small
Medium
Large
p
Roughness
Strain
Pitch
Loudness
Breathiness
Severity
25.32
30.74
40.31
<0.0001
31.11
35.18
48.24
<0.0001
14.62
22.32
27.22
0.0072
3.00
8.11
–4.19
0.0165
20.94
23.60
27.59
0.1847
29.68
37.21
49.04
<0.0001
CAPE-V — Consensus Auditory-Perceptual Evaluation of Voice.
Nuss et al, Vocal Nodule Size & Voice Quality
135
Fig 3. Mean values of perceptual characteristics by nodule type (sessile versus discrete).
RESULTS
A total of 145 patients met the selection criteria.
The distribution of patients in this study is displayed
in Fig 1. There was an approximately 2-to-1 maleto-female ratio, with 66% male and 34% female. The
preschool age group, 2 to 5.9 years of age, made up
31% of patients in this study. The school-age group,
6.0 to 11.9 years of age, included 61% of the study
population. Teenagers, 12 to 18 years of age, were
just 8% of the study population. Overall, small nodules were noted in 23% of patients, moderate nodules in 39%, and large nodules in 37%.
Fig 4. Mean values of perceptual characteristics according to age.
ated with roughness (p = 0.0286) and overall severity (p = 0.0349). The group 6 to 11.9 years of age
had the smallest disturbance, and the groups 2 to 5.9
years of age and more than 12 years of age had very
similar levels of disturbance, as seen in Fig 4 and
Table 3.
Gender was not found to be a significant factor
associated with any perceptual characteristics.
Univariate analysis showed a statistically significant association between nodule size and the perceptual measures of roughness, strain, pitch, loudness,
and overall severity. With the exception of loudness,
as vocal fold nodule size increased, the mean value
of perceptual characteristics became larger. The association of each perceptual voice characteristic and
vocal fold nodule size is shown in Fig 2. Table 1
lists the value of means and p values according to
the F-test.
In a multivariate analysis of overall severity of
voice disturbance, vocal fold nodule size and patient
age were found to be significant independent variables, with p values of less than 0.0001 and 0.0023,
respectively. Interaction between these two variables
was not significant. After adjusting for age, the difference of mean severity between nodule sizes large
and small was 20.19 (p < 0.0001), that between sizes large and medium was 13.6 (p < 0.0001), and
that between sizes medium and small was 6.59 (p =
0.1513). Figure 5 displays the estimated means for
overall severity of voice disturbance based on multivariate analysis of the independent variables age and
vocal fold nodule size. Table 4 includes numerical
data of the same.
Nodule type (sessile versus discrete) was also
analyzed as an independent factor for perceptual
characteristics. Analysis of results demonstrated
that the mean value of perceptual characteristics was
larger when the nodule contour was discrete rather
than sessile. However, statistical significance was
achieved only with the characteristic of roughness.
Figure 3 and Table 2 summarize the means and p
values of perceptual characteristics by nodule type.
DISCUSSION
The ability to assess vocal fold nodules in children has been limited in the past by the lack of a val-
Age was noted to be a significant factor associ-
TABLE 2. MEAN SCORE OF PERCEPTUAL CHARACTERISTICS, BASED ON CAPE-V SCALE, BY NODULE TYPE
Nodule Type
Sessile
Discrete
p
Roughness
Strain
Pitch
Loudness
Breathiness
Severity
31.66
38.55
0.0392
37.83
43.77
0.1638
22.01
23.60
0.6768
1.97
3.73
0.7565
23.61
27.63
0.7565
38.77
43.97
0.1653
136
Nuss et al, Vocal Nodule Size & Voice Quality
TABLE 3. MEAN SCORE OF PERCEPTUAL CHARACTERISTICS, BASED ON CAPE-V SCALE, BY AGE
Age Group
2-5.9 y
6-11.9 y
>12 y
p
Roughness
Strain
Pitch
Loudness
Breathiness
Severity
37.91
30.28
36.09
0.0286
44.86
36.83
41.67
0.2614
25.59
20.51
24.00
0.3151
8.52
–0.67
1.92
0.0905
25.64
22.86
31.92
0.1996
44.93
36.80
44.45
0.0349
idated scale to report the size of the nodule, as well
as the absence of a validated scale to rate the perceptual characteristics of voice quality. In the absence
of accepted valid scales, reports of voice quality in
children with vocal fold nodules have been difficult
to interpret and sometimes contradictory.9-13
Presently, there are excellent tools both to report
the size of vocal fold nodules in children and to describe the perceptual characteristics of the child’s
voice. A validated scale used to rate the size of pediatric vocal fold nodules has been created, and is
proving useful in a clinical context, as well as in
clinical research.7 The CAPE-V scale has been successfully used in the pediatric population, providing
a significant improvement in our ability to describe
a child’s hoarse voice quality.
This study was undertaken to better understand
whether there indeed is any relationship between the
size of vocal fold nodules in children and the associated voice disturbance as perceived by an experienced speech and language pathologist. A previous
study failed to demonstrate any significant correlation, but that study may have been flawed by the
lack of a validated instrument for assessing voice
quality.12
The important finding in the present study is that
there is a statistically significant association between
nodule size and the perceptual measures of roughness, strain, pitch, and overall severity. As vocal fold
nodule size increases, the mean value of these perceptual characteristics becomes larger. In summary,
size matters. The contour of the nodule (sessile or
discrete) was not generally found to be a significant
factor associated with voice quality. The age of the
child was noted to be significant for voice qualities of roughness and overall severity; the youngest
age group (2 to 5.9 years) and the oldest age group
(more than 12 years) were more severely affected by
the presence of nodules. Interestingly, gender was
not found to be a significant independent risk factor
for voice quality.
The results of this study may be useful in a clinical context, as following a child’s voice quality by
use of the CAPE-V scale may be a reliable guide to
the size of the underlying vocal fold nodules. Improvements in a child’s voice quality would suggest
a decrease in the size of the nodules. A worsening
voice quality may indicate increasing size of vocal
fold nodules, or the development of another factor
that is contributing to the voice disturbance. Clinical
observations of a child’s larynx, as obtained during
fiberoptic videolaryngoscopy, will continue to be the
gold standard of the health and condition of the vocal folds. However, there are frequent occasions in
which we would like to reassure an anxious parent
that their child’s vocal fold nodules are improving —
without the need for a repeat fiberoptic examination.
The present report suggests that perceptual assessment of a child’s voice quality, as performed longitudinally on several occasions, is a reliable guide to
the size of previously diagnosed nodules. Perceptual
assessment of voice quality is performed by an experienced speech and language pathologist, and may
be used in conjunction with patient-based surveys to
improve our ability to describe voice disorders and
offer optimal management.14 This report indicates
that perceptual assessment using the CAPE-V scale
in children offers an indirect means to follow vocal
fold nodule size.
TABLE 4. ESTIMATED MEAN SCORE FOR OVERALL
SEVERITY OF VOICE DISTURBANCE (AS RATED
WITH CAPE-V SCALE) BASED ON MULTIVARIATE
ANALYSIS OF AGE AND VOCAL FOLD NODULE SIZE
Fig 5. Estimated means for overall severity of voice disturbance based on multivariate analysis of independent
variables age and vocal fold nodule size.
Nodule Size
Small
Medium
Large
Age 2-5.9
Years
36.29
42.88
56.48
Age 6-11.9
Years
26.01
32.60
46.20
Age >12
Years
34.08
40.67
54.27
Nuss et al, Vocal Nodule Size & Voice Quality
Clinical research of pediatric voice disorders requires an ongoing emphasis on the use of validated
instruments. The CAPE-V scale and a previously
published scale of vocal fold nodule sizes7 are important tools to assist researchers in describing the
effectiveness of various therapies and interventions.
CONCLUSIONS
The size of vocal fold nodules in children is noted
137
to have a significant statistical correlation with perceptual qualities, including overall severity of voice
disturbance, roughness, strain, pitch, and loudness.
With the exception of loudness, as vocal fold nodule
size increases, the mean value of perceptual characteristics becomes larger. The present report indicates
that the perceptual assessment of a child’s voice
quality using the CAPE-V scale offers an indirect
means to follow vocal fold nodule size.
REFERENCES
1. Akif Kiliç M, Okur E, Yildirim I, Güzelsoy S. The prevalence of vocal fold nodules in school age children. Int J Pediatr
Otorhinolaryngol 2004;68:409-12.
evaluation of voice: development of a standardized clinical protocol. Am J Speech Lang Pathol 2009;18:124-32.
2. Hirschberg J, Dejonckere PH, Hirano M, Mori K,
Schultz-Coulon HJ, Vrticka K. Voice disorders in children. Int J
Pediatr Otorhinolaryngol 1995;32(suppl):S109-S125.
9. Lee L, Stemple JC, Glaze L, Kelchner LN. Quick screen
for voice and supplementary documents for identifying pediatric voice disorders. Lang Speech Hear Serv Sch 2004;35:30819.
3. De Bodt MS, Ketelslagers K, Peeters T, et al. Evolution
of vocal fold nodules from childhood to adolescence. J Voice
2007;21:151-6.
10. Niedzielska G. Acoustic analysis in the diagnosis of voice
disorders in children. Int J Pediatr Otorhinolaryngol 2001;57:
189-93.
4. Gray SD, Hammond E, Hanson DF. Benign pathologic
responses of the larynx. Ann Otol Rhinol Laryngol 1995;104:
13-8.
11. Niedzielska G, Glijer E, Niedzielska A. Acoustic analysis of voice in children with noduli vocales. Int J Pediatr Otorhinolaryngol 2001;60:119-22.
5. Hooper CR. Treatment of voice disorders in children.
Lang Speech Hear Serv Sch 2004;35:320-6.
12. Shah RK, Engel SH, Choi SS. Relationship between
voice quality and vocal nodule size. Otolaryngol Head Neck
Surg 2008;139:723-6.
6. McMurray JS. Disorders of phonation in children. Pediatr Clin North Am 2003;50:363-80.
7. Shah RK, Feldman HA, Nuss RC. A grading scale for pediatric vocal fold nodules. Otolaryngol Head Neck Surg 2007;
136:193-7.
8. Kempster GB, Gerratt BR, Verdolini Abbott K, Barkmeier-Kraemer J, Hillman RE. Consensus auditory-perceptual
13. Shah RK, Woodnorth GH, Glynn A, Nuss RC. Pediatric
vocal nodules: correlation with perceptual voice analysis. Int J
Pediatr Otorhinolaryngol 2005;69:903-9.
14. Karnell MP, Melton SD, Childes JM, Coleman TC, Dailey SA, Hoffman HT. Reliability of clinician-based (GRBAS
and CAPE-V) and patient-based (V-RQOL and IPVI) documentation of voice disorders. J Voice 2007;21:576-90.
The Management of Postintubation Phonatory Insufficiency
Lindsey C. Arviso, MD; Adam M. Klein, MD; Michael M. Johns III, MD
Objectives: Prolonged intubation may lead to medial arytenoid cartilage erosion and cricoarytenoid joint scarring with
subsequent glottic insufficiency. This has been referred to as post-intubation phonatory insufficiency (PIPI). Reports on
treatment outcomes for this condition are lacking.
Methods: A single institution retrospective chart review from January 2007 to the present was performed for PIPI diagnosis. Data were collected for injury, symptoms, diagnosis, interventions, and outcomes.
Results: Four patients with PIPI underwent treatment to improve voice. Patient 1 underwent injection augmentation of
the anatomic deficiency as well as the ipsilateral true vocal fold. Patient 2 had a revision thyroplasty and subsequently 2
injections of the posterior defect and true vocal fold. Patient 3 was treated with thyroplasty and arytenoid adduction. Patient
4 underwent thyroplasty with adduction arytenopexy. Limited to no phonatory improvement was achieved in any case.
Conclusions: The management of patients with PIPI is difficult. Currently, no reliable means are available to restore
cartilaginous defects in the respiratory glottis. Patients with this condition should be counseled as to the difficult nature
of treatment.
Analysis of Vocal Fold Injection Amount:
Association With Diagnosis and Outcome
VyVy N. Young, MD; Libby J. Smith, DO
Objectives: We reviewed a large database of vocal fold injection (VFI) patients, to determine a correlation between
amount injected, diagnosis, and outcome.
Methods: Retrospective review of surgical and clinical databases was performed on all patients undergoing VFI between
July 2005 and August 2009.
Results: 335 patients were identified. Patients undergoing unilateral VFI for paralysis or bilateral VFI for atrophy who
had pre- and post-operative Voice Handicap Index-10 (VHI) scores (n = 102) were analyzed. The average amount injected
was 0.58 cc for paralysis and 0.78 cc for atrophy. The average amounts injected for paralysis patients were 0.67 cc for
“great,” 0.69 cc for “good,” 0.49 cc for “fair,” and 0.56 cc for “poor” outcomes. The average amounts injected for atrophy
patients were 0.8 cc for “great,” 0.63 cc for “good,” 0.88 cc for “fair,” and 0.90 cc for “poor” outcomes. Comparison was
made between patients with more versus less favorable outcomes. None of these differences were statistically significant
(p values > 0.168).
Conclusions: Vocal fold injection is increasingly used for management of atrophy and paralysis. However, the “optimal”
amount of material to inject remains undefined. Presently, there are no reports in the literature that numerically delineate
this quantity, forcing surgeons to rely on their own best judgment. This is the first paper which attempts to define numerically an appropriate amount of injection material for atrophy and paralysis patients, and to try to correlate the amount of
injected material with outcome. The current study demonstrates no significant difference between injected amounts based
on diagnosis or outcome, suggesting that the optimal quantity is based on factors other than simply diagnosis.
138
Glidescope-Assisted Medialization Laryngoplasty:
A Novel Technique
Roberta Lima, MD; Nazaneen Grant, MD
Objectives: Injection medialization laryngoplasty can be performed under direct laryngoscopy or local anesthesia with a
flexible scope. While sufficient in most cases, these techniques are limited in certain patient populations with anatomical
limitations to laryngeal exposure, including the patient whose C-spine precludes extension, the irradiated neck, and the
patient who cannot tolerate an office procedure. Our objective is to describe a novel technique of injection with direct
visualization under general anesthesia utilizing video-assisted blade laryngoscopy for vocal fold medialization in a select
population.
Methods: In a case series of three patients, the technique, laryngeal exposure, and duration of the procedure were explored.
All three patients had dysphonia with glottic insufficiency and refused awake medialization under local anesthesia. Two
had cervical spine abnormalities that precluded full neck extension, and one had severe radiation fibrosis of the neck.
Calcium hydroxylapatite paste was injected with a flexible transoral injection needle.
Results: The Glidescope, a curved laryngoscopy blade with an attached video monitor, produces a sufficient view of
the larynx without the need for a direct line of vision from the examiner to the larynx. In two patients, video-assisted
laryngoscopy provided excellent visualization for successful injection medialization. In one patient, the visualization of
the anterior vocal folds was not adequate and a flexible bronchoscope was additionally used, resulting in a successful
medialization. For two patients, the time of the procedure was less than with traditional approaches.
Conclusions: Video-assisted blade laryngoscopy for vocal fold medialization in patients with difficult laryngeal exposure
is a novel and effective technique.
Laryngeal Findings in Occluded Versus Unoccluded
Ostia Model of Rhinosinusitis
Ankona Ghosh; Marcelo B. Antunes, MD; Joel Guss, MD; Fenella Long, PhD;
Noam A. Cohen, MD, PhD; Natasha Mirza, MD
Rhinosinusitis and laryngitis are often found to coexist. Patients with rhinosinusitis exhibit a range of laryngeal complaints,
including hoarseness, persistent cough, and globus sensation. The purpose of this study was to determine if there was
evidence of laryngeal inflammation in a rabbit model of sinusitis. Acute sinusitis was induced in 2 sets of rabbits. In 11
rabbits, the maxillary ostium was occluded and a bacterial inoculum was placed in the sinus to induce sinusitis. After
1 week, the sinus was unoccluded in 5 rabbits, resulting in purulent rhinorrhea that was allowed to persist for 1 week
prior to sacrificing the animal (occluded). The remaining 6 rabbits were sacrificed after 1 week without unocclusion
of the inoculated sinus (unoccluded). Three rabbits served as normal controls. Inflammation in each larynx (using H &
E–stained slides from tissue at the levels of supraglottis, glottis, and subglottis) was scored by a blinded pathologist on
a scale of 1 to 17. There was no statistically significant difference in the inflammatory score between occluded, unoccluded, and control rabbit larynges (p = 0.20). There was, however, a trend toward a higher inflammatory score in the
unoccluded group. One week of acute sinusitis with or without purulent rhinorrhea is not associated with significant
laryngitis in rabbits. However, higher inflammatory scores in the unoccluded group versus the occluded group may
indicate that postnasal discharge has a direct irritative effect on the larynx. While the model is limited, it provides the
initial steps in studying the relationship between rhinosinusitis and laryngitis.
139
Narrowband Imaging and High-Definition Television for
Detection, Definition, and Surveillance of Head and Neck Cancer:
A Prospective Study
Cesare Piazza, MD; Daniela Cocco, MD; Francesca Del Bon, MD;
Stefano Mangili, MD; Luigi de Benedetto, MD; Giorgio Peretti, MD
Objectives: Narrowband imaging (NBI) is an optical technique in which a filtered light enhances the neoangiogenic pattern of neoplasms. Its accuracy is implemented by a high-definition television (HDTV) camera. The aim of the study was
to evaluate the diagnostic gain in the pre-, intra-, and postoperative evaluation of head and neck cancer (HNC).
Methods: 419 patients who had squamous HNC of the nasopharynx, oropharynx, hypopharynx, oral cavity, or larynx or
were previously treated for it were prospectively evaluated by white light (WL) and NBI ± HDTV between April 2007
and August 2009. The patients were divided into group A (157 patients submitted to pre- and intraoperative WL and NBI
endoscopy) and group B (262 subjects evaluated at least 6 months after treatment). Tumor resection was performed, taking into account NBI information. The sensitivity, specificity, positive and negative predictive values, and accuracy were
calculated.
Results: Overall, 104 of the 419 patients (25%) showed adjunctive findings with NBI and HDTV in comparison to standard
WL endoscopy. Among them, 91 (87%) received histopathologic confirmation (false-positive rate, 13%). The sensitivity,
specificity, positive and negative predictive values, and accuracy were 98%, 83%, 96%, 98%, and 91%.
Conclusions: NBI and HDTV showed its value in better defining tumor extension (upstaging of 51 neoplasms), detecting 5 synchronous lesions, evaluating an incomplete response to radiotherapy before planned neck dissection in 3 cases,
and identifying 2 unknown primaries. NBI in the post-treatment setting precociously detected 26 recurrences and 4
metachronous tumors.
Optimizing the Surgical Creation of a Supraglottal
Sound Source: Theory and a Case Study
Sid Khosla, MD; Shanmugam Murugappan, PhD; Bernice Klaben, PhD
Objectives: In the classic source-filter theory, the vibrating vocal folds act as the source of sound and the vocal tract
acts as the filter. After reconstructive surgery for conditions such as laryngeal stenosis, trauma, or cancer, the vocal folds
sometimes do not effectively vibrate. As a compensatory mechanism, vibration of supraglottal structures will be used as
the primary source of sound. The resulting voice is often harsh, aperiodic, not loud enough, and difficult to understand.
Methods: Our work in an animal model has shown that vortices are important for producing a loud, intelligible voice;
we have also discovered some of the necessary biomechanical conditions for optimal vortex production during phonation. These biomechanical conditions will be discussed, along with a description of how these conditions were met in
the laryngeal reconstruction of a patient. This patient suffered a complete crush injury of his thyroid, and two potentially
vibrating structures were built on the medial surface of the arytenoids. After 2 months, endoscopic surgery was then done
to create the necessary conditions mentioned above.
Results: Stroboscopic and high-speed videography show relatively periodic, symmetric vibrations of the new sound
source. The vocal intensity range is from 40 to 100 dB. Quality-of-life scores and jitter and shimmer are within the normal
range.
Conclusions: In one patient, we show how our understanding of vortex production, developed in an animal model, can
be used to create an optimal supraglottal vibrating sound source.
140
Primary TEP Placement in Patients With Laryngopharyngeal
Free Tissue Reconstruction and Salivary Bypass Tube Placement
Vasu Divi, MD; Derrick T. Lin, MD; Kevin Emerick, MD; Daniel G. Deschler, MD
Objectives: We examined the feasibility and advantages of primary tracheoesophageal puncture (TEP) with intraoperative placement of the voice prosthesis for patients undergoing laryngopharyngectomy requiring free tissue reconstruction
and salivary bypass tube placement.
Methods: After Institutional Review Board approval, a retrospective chart review was completed of all cases of primary
tracheoesophageal prosthesis placement completed at the time of laryngopharyngectomy and free tissue reconstruction
of the pharynx.
Results: Six patients were identified; 4 underwent total laryngopharyngectomy and 2 underwent total laryngectomy
with partial pharyngectomy. Three patients received preoperative full-dose radiotherapy to the larynx, and 1 received
full-dose irradiation for an oral cavity primary. Radial forearm free flap reconstruction was performed in 5 patients, and
an anterolateral thigh flap in 1. A salivary bypass tube was placed in all cases. All patients had a 20F Indwelling BlomSinger prosthesis (InHealth Technologies, Carpinteria, California) placed. No complications were noted with intraoperative prosthesis placement. No prostheses were dislodged in the postoperative period. Three of the 6 subjects had initial
success with tracheoesophageal voice production. One patient required removal of the TEP postoperatively for feeding
tube placement. The prosthesis was replaced 1 month later with good voice restoration. One patient died of disease before
voice evaluation, and 1 patient was lost to follow-up. At 6 months, 4 patients available for evaluation had successful voice
outcomes and 3 were disease-free.
Conclusions: This study demonstrates the effectiveness of voice prosthesis placement at the time of primary TEP associated with free tissue reconstruction of a laryngopharyngeal defect.
Development of the Dyspnea Severity Index–10
Jackie Gartner-Schmidt, PhD, SLP; Adrianna Shembel, BS, MA; Priya Krishna, MD;
Rita Hersan, CCC-SLP; Thomas Zullo, PhD; Clark A. Rosen, MD
Paradoxical vocal fold motion disorder (PVFMD) is a condition in which the vocal folds adduct involuntarily during
inspiration and sometimes during exhalation. The purpose of this study was to develop a dyspnea severity index that can
be used as a clinical tool to 1) estimate perceived severity and 2) evaluate possible treatment outcomes in patients with
dyspnea related to the upper airway. A clinical consensus with laryngologists and speech-language pathologists was used
to develop a list of clinically significant questions/items. Two hundred consecutive patients who either had a complaint of
cough and/or dyspnea filled out the original instrument. Principal component analyses and item analyses were performed.
Factor analysis was performed, confirming that the majority of the questions were related to a single factor. Another
clinical consensus was held to reduce the questionnaire to 10 questions each, and it was re-run for reliability. Cronbach’s
alpha coefficient demonstrated excellent internal reliability. The Dyspnea Severity Index–10 allows clinicians to quantify
patients’ perception of the problems related to dyspnea and may allow for treatment results evaluation in the future.
141
Pepsin, at pH 7, in Nonacidic Laryngopharyngeal Refluxate,
Significantly Alters the Expression of Multiple Genes
Implicated in Carcinogenesis
Nikki Johnston, PhD
Objectives: The role of reflux as a causative agent in laryngeal cancer is controversial. Many clinical studies strongly
support an association between reflux and laryngeal cancer; however, it is difficult to prove causality. The objective of this
study was to investigate the effect of pepsin exposure on the expression of 84 genes implicated in carcinogenesis.
Methods: Human hypopharyngeal epithelial FaDu cells were incubated with or without 0.1 mg/mL human pepsin at pH
7, 37°C, overnight. The expression of 84 genes implicated in carcinogenesis was analyzed via RT2 qPCR array.
Results: The expression level of 27 genes implicated in carcinogenesis was significantly altered in human hypopharyngeal
epithelial cells exposed to pepsin at neutral pH relative to control cells (>1.5-fold change in gene expression; p < 0.05).
Genes involved in cell cycle control, DNA damage repair, apoptosis and cell senescence, signal transduction molecules
and transcription factors, adhesion, angiogenesis, and invasion and metastasis were significantly affected by pepsin at pH
7 compared to pH 7 control.
Conclusions: Reflux of pepsin, even at nonacidic pH, may result in neoplastic changes in the aerodigestive tract.
The Role of the Modified Barium Swallow Study and
Esophagram in Patients With GERD/Globus Sensation
Jayme R. Dowdall, MD; Tom Willis, MD; Adam J. Folbe, MD; James P. Dworkin, PhD
Objectives: Globus sensation is a common benign finding that is often associated with frequent throat clearing and is
commonly a result of laryngopharyngeal reflux. We aimed to further examine the role of the modified barium swallow
study (MBSS) with esophagram. We hypothesized that a radiographic swallow study does not add diagnostically significant
information in the investigation of globus sensation.
Methods: We performed a retrospective chart review of patients presenting to Karmanos Cancer Institute with a chief
complaint of globus sensation between 2000 and 2009 who underwent both MBSS and esophagram. IRB approval was
obtained.
Results: Of the 380 patients who underwent MBSS, only 68 patients were eligible for this study. Over 70% of patients were
on reflux medicines; 81% of the MBSS studies were completely normal; 62% of the esophagram results were completely
normal; and 18% of patients were noted to have hiatal hernia, 10% with mild reflux. Esophagoscopy was performed in 45%
of patients, of which the findings of 35% were normal. One patient initially had a normal EDG and then was subsequently
diagnosed with gastric cancer. Hiatal hernia was noted in 45%. Fifty-nine percent of patients underwent CT scans of the
neck with IV contrast, of which 67% had minor findings.
Conclusions: Positive findings are often benign and can be treated with reflux medications. Esophagoscopy often showed
normal findings and was most sensitive for hiatal hernia. No hypopharyngeal cancer was noted. Therefore, MBSS and
esophagram for patients with globus sensation is most often negative and does not add additional diagnostic information.
142
Effect of Different Angiolytic Lasers Upon Resolution of
Submucosal Hematoma in an Animal Model
Daniel Novakovic, MD; Joanna D’Elia, MD; Andrew Blitzer, MD; Milton Waner, MD
Objectives: Hematoma of the vocal fold is traditionally treated with a period of voice rest, on the order of weeks, to allow
natural resolution. This downtime may have significant impact upon the professional voice user. A number of hemoglobinavid lasers exist on the market today. This study was designed to examine whether treatment with such lasers will accelerate
resolution of submucosal hematoma.
Methods: This was a prospective, blinded, controlled interventional animal study. Venous blood was drawn from 6 white
rabbits and was used to create an array of 0.05 mL submucosal hematomas in the buccal cavities of each animal. Laser
energy from one of three different lasers (532 nm KTP, 532 nm diode, or pulsed dye laser) was applied to each of the test
hematomas at varying energy levels. Lesions were photographed before intervention and then after intervention at days 0,
2, 4, 6, 8, 10, 12, and 14. Animals were sacrificed on day 14, and the hematoma grid was assessed by a blinded pathologist for evaluation of hematoma resolution and collateral tissue damage. Photographs were assessed for color density and
determination of time to resolution.
Results/Conclusions: We present an animal model of submucosal hematoma and discuss our findings with respect to
resolution of hematoma treated with hemoglobin-avid lasers.
Characterization of Mesenchymal Stem Cells From Human
Vocal Fold Fibroblasts
Summer E. Hanson, MD; Jaehyup Kim, MD; Beatriz H. Quinchia Johnson, DDS, PhD;
Melissa Breunig; Bridget Bradley; Peiman Hematti, MD; Susan L. Thibeault, PhD
Objectives: Mesenchymal stem cells (MSCs), originally isolated from bone marrow, are fibroblast-looking cells that are
now assumed to be present in the stromal component of many tissues. MSCs are characterized by a certain set of criteria, including their growth culture characteristics, a combination of cell surface markers, and the ability to differentiate
along multiple mesenchymal tissue lineages. We hypothesized that human vocal fold fibroblasts (hVFF) isolated from
the lamina propria meet the criteria established to define MSCs and are functionally similar to MSCs derived from bone
marrow (BM) and adipose tissue (AT).
Methods: hVFF were previously derived from human vocal fold tissues. MSCs were derived from BM and AT of healthy
donors, based on their attachment to culture dishes and their morphology, and expanded in culture. Cells were analyzed
for standard cell surface markers identified on BM-derived MSCs as well as the ability to differentiate into cells of
mesenchymal lineage, ie, fat, bone, and cartilage. We investigated the immunophenotype of these cells before and after
interferon-γ (INF-γ) stimulation.
Results: hVFF displayed cell surface markers and multipotent differentiation capacity characteristic of MSCs. Furthermore,
these cells exhibited similar patterns of expression of HLA and co-stimulatory molecules after stimulation with INF-γ
compared to MSCs derived from BM and AT.
Conclusions: Based on our findings, hVFF derived from lamina propria have the same cell surface markers, immunophenotypic characteristics, and differentiation potential as BM- and AT-derived MSCs. We propose that VF fibroblasts
are MSCs resident in the vocal fold lamina propria.
143
Utility of Diagnostic Testing in Adults With
Idiopathic Subglottic Stenosis
Melissa McCarty Statham, MD; Yash A. Patil, MD; Allesandro de Alarcon, MD;
Ravindra G. Elluru, MD, PhD; Meredith E. Tabangin, MPH; Michael W. Bowen, PAC;
Thomas K. Chung, BS; Michael J. Rutter, MD, FRACS
Objectives: We describe a cohort with idiopathic subglottic stenosis (ISGS) and examine the utility of investigative studies
performed to diagnose and guide treatment of ISGS-associated airway obstruction.
Methods: We performed a retrospective review.
Results: A total of 36 patients with ISGS were treated from 1999 to 2009. All patients were female, with a mean age
of 43.3 years (19.9 to 63). All patients exhibited focal subglottic stenotic lesions, with the average size of the subglottis
accommodating a 4.5 endotracheal tube on initial evaluation and 64% (n = 23) of subjects demonstrating a Cotton-Myer
grade III subglottic stenosis. Seven of the 36 patients (19.4%) reported worsening of their symptoms during the second
trimester of pregnancy. The medical comorbidities examined included hypertension (27.8%), reflux (22.2%), asthma
(16.7%), diabetes mellitus (8.3%), obstructive sleep apnea (8.3%), and rheumatologic disorders (none). The average
body mass index (BMI) approached obesity (29.9), and only 27.8% of patients had a BMI in the normal range. The mean
number of diagnostic tests performed in this cohort was 10.8 ± 3.2 (range, 4 to 18). Erythrocyte sedimentation rate or
C-reactive protein was elevated in 30.6% (11/36). None of the patients demonstrated positive serology for circulating
or perinuclear anti-neutrophil cytoplasmic antibodies (c-ANCA, p-ANCA). Other than subcentimeter thyroid nodules,
computed tomography of the neck demonstrated focal subglottic narrowing in studied patients.
Conclusions: ISGS is a diagnosis of exclusion in the evaluation of patients with subglottic stenosis. In this cohort, an
extensive radiographic and serologic evaluation was not predictive in guiding treatment of ISGS in the absence of systemic
comorbidities or findings suggestive of a rheumatologic process.
A Challenging Case: Intubation Through a Foreign Body
and Foreign Body Removal From the Larynx
Vartan A. Mardirossian, MD; Timothy Anderson, MD; Joyce Colton-House, MD
The patient was a 70-year-old man with chronic dysphagia and altered mental status after a fall who was seen in a tertiary
care center clinic for sudden significant worsening of dysphagia, dysphonia, and drooling over the past several hours. The
fiberoptic examination of the larynx showed that a complete superior denture was lodged around the epiglottis with the
teeth in the valleculae and the metallic frame and hooks against the posterior wall of the hypopharynx. A single attempt
was made to remove it in the clinic, but this was unsuccessful. During the visit and in the hour to follow, the patient developed signs of mild to moderate airway obstruction but maintained good oxygenation. He was brought urgently to the
OR, where he was fiberoptically intubated. Given the particular position and the conformation of the foreign body, the
intubation was performed through it. Subsequently, with the help of endoscopic forceps, the denture was slid upward and
out along the endotracheal tube; once the denture was out of the patient’s mouth, the anesthesia circuit had to be interrupted
in order to complete its removal from the end of the endotracheal tube. Despite some mild edema of the epiglottis and the
aryepiglottic folds caused by the prolonged presence of the foreign body in the upper airway, the patient was successfully
extubated and transferred to the postoperative care unit and then to the floor in stable condition.
144
Calcium Oxalate Crystals Decode the Pulmonary
Rhizopuzzle in a Pulmonary Abscess
Christopher G. Tang, MD; Christina C. Goette, MD; John Lin, MD;
Valerie Ng, MD, PhD; Alden H. Harken, MD
Objectives: We emphasize the calcium oxalate crystal signature of potentially devastating craniofacial and pulmonary
mucormycosis.
Methods: We examined a patient status post treatment of a necrotizing pulmonary abscess with microbiological and
pathological follow-up.
Results: The patient was a 69-year-old man admitted for pneumonia who developed a necrotizing pulmonary abscess.
Tracheobronchial cultures initially grew out large septate hyphae suggestive of aspergillus. A left anterior thoracoplasty
with a pectoral and intercostal muscle flap was performed. Calcium oxalate crystals were present in the abscess fluid suggestive for an infection by Aspergillus niger. However, the rib resection grew out aseptate hyphae suggestive of Rhizopus
microsporus (class Zygomycetes), which was also confirmed by culture.
Conclusions: Mucormycosis, a devastating fungal infection that often causes necrotizing abscesses of the sinuses and
brain in rhinocerebral manifestations, can sometimes cause pulmonary infections in immunocompromised individuals.
Rarely do these Zygomycetes produce calcium oxalate crystals, which are essentially pathognomonic for necrotizing
aspergillosis, particularly Aspergillus niger. There have only been 3 documented cases of pulmonary mucormycosis with
evidence of calcium oxalate crystals, 2 of which were in patients with concomitant oxalosis. This documented case of
pulmonary mucormycosis emphasizes that calcium oxalate crystals can be found in patients with fungal infections other
than aspergillus.
In-Office Unsedated Transnasal Bronchoscopy for
Removal of an Airway Foreign Body
Drew Plonk, MD; S. Carter Wright, Jr, MD; J. Whit Mims, MD; Catherine J. Rees, MD
Objectives: We describe the use of a 5.1-mm distal chip transnasal endoscope in the unsedated office setting to remove
an airway foreign body.
Methods: We report the single case of a 90-year-old woman with a history of severe COPD requiring home oxygen who
presented to the otolaryngology clinic complaining of persistent cough that began after “choking” on a piece of corn the
previous night.
Results: The patient was not in distress, and her comorbidities made general anesthesia a high risk; therefore, the decision
was made to proceed with unsedated, in-office bronchoscopy for diagnosis and possible intervention. After the application
of topical anesthesia, the scope was advanced into the airway and a single corn kernel was identified in the right middle
lobe distal bronchi. A combination of cup forceps and the suction port of the endoscope were used to withdraw the corn
kernel from the right lung into the nasopharynx. The corn kernel was positioned deliberately into the pyriform sinus, and
the foreign body was swallowed under direct visualization. The patient tolerated the procedure well.
Conclusions: In-office unsedated bronchoscopy is an additional option when considering the removal of an airway foreign
body in the appropriate patient and setting.
145
Endoscopic Management of a Penetrating Foreign Body
in the Soft Tissue of the Retropharynx
Margaret L. Skinner, MD; Mark Volk, DMD, MD
Objectives: We review the challenges of endoscopic treatment of a foreign body embedded in the soft tissue of the retropharynx and the unique management techniques employed.
Methods: A patient presented to a tertiary care children’s hospital, intubated and sedated, after unproductive endoscopic
exploration for a metallic foreign body. Plain film images and intraoperative fluoroscopy confirmed an aspirated straight
sewing needle to be localized to the soft tissues of the retropharyngeal and parapharyngeal space at the level of the hypopharynx. Due to concern that traditional neck exploration would expose the surgical team to high risk of needle stick
injury, endoscopic neck exploration was undertaken. Specific challenges included management of the endotracheal tube
(ETT), localization of the foreign body below the mucosal surface, and endoscopic dissection of the soft tissue.
Results: Nontraditional instrumentation and intraoperative fluoroscopy were used to remove an aspirated straight needle
from the soft tissues of the retropharyngeal and parapharyngeal space by endoscopic technique, alleviating the need for
open neck exploration.
Conclusions: Compared with traditional suspension laryngoscopy, the Crowe-Davis mouth gag and Draffin Rod suspension provided excellent exposure of the hypopharynx and retropharyngeal space with adequate clearance for intraoperative
C-arm fluoroscopy while controlling the ETT. Intraoperative fluoroscopy localized the foreign body and was essential
in the management of a submucosal metallic foreign body. The use of pediatric laparoscopic instruments enabled endoscopic guided dissection of the soft tissues of the retropharynx and minimally invasive extraction of the foreign body, and
minimized the risk of injury to the operative team.
Delay in the Diagnosis of Laryngeal Cleft Associated
With Tracheoesophageal Fistula
Margaret L. Skinner, MD; Stacey L. Ishman, MD; Umakanth Khatwa, MD;
Kenan Haver, MD; Rachel Rosen, MD; Craig Lillehei, MD; Reza Rahbar, MD
Objectives: We evaluated the incidence of laryngeal clefts associated with tracheoesophageal fistula (TEF) and sought to determine whether patients with TEF are more likely to have delayed diagnosis of concomitant laryngeal cleft anomalies.
Methods: A retrospective chart review of consecutive children with posterior laryngeal cleft anomalies was carried out
at a tertiary care children’s hospital between 2002 and 2008. Diagnosis of posterior laryngeal cleft was made by rigid
laryngoscopy and bronchoscopy and classified with the Benjamin-Inglis system. Diagnosis of TEF was determined by
review of the medical records. Significance was evaluated with the two-tailed Mann Whitney U-test.
Results: Fifty-three patients found to have laryngeal clefts were identified. Of the 53 patients, 27 had type 1 clefts, and
26 had type 2 clefts. The overall incidence of TEF was 15.0% (N = 8). The mean age at laryngeal cleft diagnosis was 2.98
years. For patients without TEF, the mean age at diagnosis was 2.35 years (3 months to 15 years), compared with 6.84
years (1 to 17 years) for patients with TEF (p = 0.0144).
Conclusions: The coincidence of TEF and laryngeal cleft in this series is consistent with that reported in the literature.
Cases of TEF are diagnosed and treated for laryngeal cleft anomalies later than cases without TEF (p = 0.0144). Because
of the similarity of symptoms, the diagnosis of laryngeal cleft may be delayed in patients with TEF, and screening for
laryngeal clefts should be considered in children with persistent symptoms after TEF repair.
146
Histopathologic Investigations of the Unphonated
Human Child Vocal Fold Mucosa
Kiminori Sato, MD; Hirohito Umeno, MD; Tadashi Nakashima, MD;
Satoshi Nonaka, MD; Yasuaki Harabuchi, MD
Objectives: Stellate cells (SCs) in the human maculae flavae (MFe) located at both ends of the human vocal fold mucosa
are inferred to be involved in the metabolism of extracellular matrices (EMs). The MFe are considered to be an important
structure in the growth and development of the human vocal fold mucosa. It is also hypothesized that the tensions caused
by phonation (vocal fold vibration) after birth stimulate SCs to accelerate production of EMs and form the layered structure. If the hypotheses are correct, some morphological differences should be detected between normal and unphonated
vocal fold mucosae.
Methods: Vocal fold mucosae, which have remained unphonated since birth, of 2 children (7 and 12 years old) with
cerebral palsy were investigated by light and electron microscopy and compared with those of normal subjects.
Results: The vocal fold mucosae (including MFe) were hypoplastic and rudimentary and did not have a vocal ligament,
Reinke’s space, or a layered structure. The lamina propria appeared as a uniform structure. Some SCs in the MFe showed
degeneration, and not many vesicles were present at the periphery of the cytoplasm. The SCs synthesized fewer EMs,
such as fibrous protein and glycosaminoglycan. The SCs appeared to have decreased activity.
Conclusions: The present study supports the hypothesis that the mechanotransduction caused by phonation after birth
stimulates SCs in the MFe to accelerate production of EMs and form the layered structure. Vocal fold vibration after birth
is an important factor in the growth and development of the human vocal fold mucosa.
147
2010 ROSTER OF MEMBERS
Honorary Members
1982
(1948)
1981
(1958)
(1980)
James P. Dudley, 1835 Franklin St, Apt 1103,
San Francisco, CA 94109-3457
Juan Carlos Arauz, Je Uriburur, 1252-3D,
Buenos Aires 1114, Argentina
John F. Daly, 56 Florence St, Hillsdale, NJ
07642-1034
Andres L. Delgado
1978
Mary D. Lekas, 129 Terrace Ave, Riverside, RI
02195-4726
1999
2008
Reza Shaker, Froedtert Memorial Lutheran
Hosp, Div of Gastroenterology, 9200 W Wisconsin Ave, Milwaukee, WI 53226
1992
(1978)
Arndt J. Duvall III, Univ Hosp, Box 396, 420
Delaware St SE, Minneapolis, MN 554550374
Peter Stradling, Apple Acre, Sampford Brett,
Taunton, Somerset TA4 4LB, England
2003
(1975)
L. Penfield Faber, 1725 W Harrison St, Rm
218, Chicago, IL 60612-3732
1980
(1954)
J. Allan Fields, 440 Royal Plaza Dr, Fort Lauderdale, FL 33301-2516
2009
(1995)
Charles N. Ford, Univ of Wisconsin Hosp
& Clinics, 600 Highland Ave, K4/714 CSC,
Madison, WI 53792-7375
1982
(1979)
Senior Members
Miguel A. Abado, J. No. 165 Vedado, Havana,
Cuba
2006
(1974)
Allan L. Abramson, PO Box 566, White Lake,
NY 12786-0566
2002
(1978)
2008
(1980)
Warren Y. Adkins, Jr, 680 Pawley Rd, Mt Pleasant, SC 29464-3566
John M. Fredrickson, 205 Dartmouth Dr, Albuquerque, NM 87106
2006
(1980)
1984
(1959)
John R. Ausband, 1510 Riverside Dr, Beaufort,
SC 29902-6487
William H. Friedman, 3023 N Ballas Rd, #5600, St Louis, MO 63131-2330
1990
(1976)
1985
(1956)
William L. Barton, 2147 Sautee Tri, Sautee
Nacoochee, GA 30571
Herman Froeb, 9834 Genessee, Ste 109, La
Jolla, CA 92037-1222
2002
(1975)
1991
(1971)
James D. Baxter, 1237 Northshore Blvd E, Apt
908, Burlington, ON L7S 2H8, Canada
Willard A. Fry, 1111 Sunset Rd, Winnetka, IL
60093
2007
(1993)
1986
(1975)
George Berci, 8700 Beverly Blvd, Apt 8215,
Los Angeles, CA 90048-1804
Edward B. Gaynor, 252 Valley Ridge Rd, Haverford, PA 19041
2009
(1995)
1999
(1987)
Hugh F. Biller, 5 E 98th St, New York, NY
10029
Jack L. Gluckman, 3 Grandin Lane, Cincinnati,
OH 45208
2005
(1993)
1999
(1989)
Donald S. Blatnik, 22972 Shady Knoll Dr,
Bonita Springs, FL 34135
Michael E. Goldman, 3618 Pebble Beach Rd,
Northbrook, IL 60062
2004
(1985)
2001
(1969)
Stanley M. Blaugrund, 787 Park Ave, New
York, NY 10021-3552
Charles W. Gross, UVA Health Sci Ctr, Dept of
Oto, Box 800713, Charlottesville, VA 229080713
2005
(1971)
Charles D. Bluestone, Children’s Hosp of Pittsburgh, Dept of Oto, 3705 Fifth Ave, Pittsburgh,
PA 15213-2583
2003
(1985)
Thomas W. Grossman, 8008 Limerick Rd, Milwaukee, WI 53223-1072
2009
(1982)
1998
(1978)
Roger Boles, Box 620203, Woodside, CA 94062
2009
(1991)
Patrick J. Bradley, 37 Lucknow Dr, Mapperley
Park, Nottingham NG3 5EU, England
Kenneth M. Grundfast, Dept of Oto, Boston
Med Ctr, D616, 88 E Newton St, Boston, MA
02118-2308
1996
(1978)
2003
(1974)
Thomas C. Calcaterra, UCLA HSC, 10833 Le
Conte Ave, Rm 62-158, Los Angeles, CA
90095
Donald B. Hawkins, 5049 El Adobe Lane, La
Crescenta, CA 91214-2105
2004
(1978)
Leonard L. Hays, 708 Summit Lakeshore Rd
NW, Olympia, WA 98502-9482
2001
(1976)
Robert W. Cantrell, 1925 Owensville Rd, Charlottesville, VA 22901
1987
(1953)
Henry J. Heimlich, Heimlich Inst, 311 Straight
St, Cincinnati, OH 45219
1992
(1974)
Francis I. Catlin, 8580 Woodway Dr, #1124,
Houston, TX 77063-2466
1996
(1974)
William R. Hudson, Duke Univ Med Ctr, PO
Box 3805, Durham, NC 27710-3805
1993
(1976)
Paul L. Chodosh
2009
(1990)
2004
(1982)
Noel L. Cohen, NYU Med Ctr, 530 First Ave,
New York, NY 10016-6402
Michael E. Johns, Emory Univ, 1440 Clifton
Rd NE, Ste 400, Atlanta, GA 30322
1998
(1975)
Robert I. Kohut, 2620 S Riverchurch Rd,
Woodleaf, NC 27054
2004
(1969)
George H. Conner, Lebanon VA Med Ctr, 2 E
High St, Lebanon, PA 17042-5454
2002
(1980)
2004
(1978)
Charles W. Cummings, 13940 Mantna Mill Rd,
Glyndon, MD 21071
2003
(1976)
1982
(1961)
Timothy L. Curran, 50 Winding Lane, Avon,
CT 06001-2624
1992
(1973)
Paul A. Kvale, Henry Ford Hosp, 2799 W
Grand Blvd, Detroit, MI 48202-2608
Louis D. Lowry, 503 Centerbridge Meadowood, Lansdale, PA 19446
George D. Lyons, 6323 West End Blvd, New
Orleans, LA 70112
148
Members
149
1998
(1989)
Anthony J. Maniglia, 4167 Palm Ln, Miami,
FL 33137
1984
(1965)
2000
(1973)
Bernard R. Marsh, 4244 Mount Carmel Rd,
Upperco, MD 21155-9557
Melvin L. Samuels, 12458 Woodthorpe Ln,
Houston, TX 77024-4136
2003
(1970)
2005
(1991)
Kenneth F. Mattucci, Northshore Oto Assoc, 11
Nome Dr, Woodbury, NY 11797-3404
David R. Sanderson, 10676 E Bella Vista Dr,
Scottsdale, AZ 85258
2003
(1990)
2003
(1979)
Gregory J. Matz, 910 N Lake Shore Dr, Apt
1119, Chicago, IL 60611
Gary L. Schechter, Oto-HNS, 825 Fairfax Ave,
No. 510, Norfolk, VA 23507-1912
1998
(1970)
1985
(1978)
Harry W. McCurdy, 6006 Dellwood Pl, Bethesda, MD 20817-3812
Joyce Schild, 853 W George, Chicago, IL
60657
1989
(1958)
2009
(1990)
W. Frederick McGuirt, Sr, Dept of Oto, Wake
Forest Med Ctr, Medical Center Blvd, WinstonSalem, NC 27157
Myron J. Shapiro, 81 Sand Spring Rd, Morristown, NJ 07960-6702
2001
(1974)
Harvey D. Silberman, 2401 Pennsylvania Ave,
Apt 8A4, Philadelphia, PA 19130-3030
10467-2404
1991
(1959)
Francis L. McNelis, 37 Baggy Wrinkle Cove,
Warren, RI 02885-4114
1990
(1963)
James T. Spencer, 329 Baker Ln Ridgemont,
Charleston, WV 25302
1984
(1964)
Harold C. Menger, 7809 Myrtle Ave, Glendale,
NY 11385-7439
1991
(1978)
Philip M. Sprinkle, 315 Hospital Dr, Ste 108,
Martinsville, VA 24112-1927
1991
(1987)
Peter J. Moloy, 607 Newcastle, Irvine, CA
92620
2006
(1980)
Harvey M. Tucker, Dept of Oto, Metrohealth,
2500 Metrohealth Dr, Cleveland, OH 44109
2003
(1980)
Willard B. Moran, Jr, 5914 Chestnut Ct,
Edmond, OK 73003-2515
1998
(1970)
1991
(1970)
Karl M. Morgenstein, 3650 N 36th Ave, Apt 70,
Hollywood, FL 33019-1931
John A. Tucker, Children’s Hosp of Philadelphia, Dept of Ped Oto, 34th & Civic Ctr Blvd,
1st Fl Wood, Philadelphia, PA 19104-4399
2000
(1978)
1984
(1965)
Harry R. Morse, 365 N Main St, Quail Hollow
Unit A4, West Lebanon, NH 03784
Donald P. Vrabec, 2010 Snydertown Rd, Danville, PA 17821-7720
1983
(1963)
2003
(1980)
Eugene N. Myers, Dept of Oto, Eye & Ear Inst,
200 Lothrop St, Ste 500, UPMC, Pittsburgh, PA
15213-2546
Duncan D. Walker, Jr, 114 Idle Hour Dr, Macon, GA 31210-4434
1993
(1969)
Paul H. Ward, 32178 Atosona Dr, PO Box 250,
Pauma Valley, CA 92061-0250
2006
(1978)
H. Bryan Neel III, 828 SW 8th St, Rochester,
MN 55902
1996
(1978)
Louis W. Welsh, 2236 Deer Path Rd, Huntingdon Valley, PA 19006-5906
1996
(1970)
Martin Norton, 1624 Cherokee Rd, Ann Arbor,
MI 48103-4449
1980
(1960)
Chester M. Weseman, 45 Oak Ridge Rd, Berkeley, CA 94705-2425
2006
(1978)
Moses Nussbaum, Beth Israel Med Ctr, 10 Union
Square E, Ste 4J, New York, NY 10003-3881
2009
(1981)
1989
(1971)
Joan O’Brien
2001
(1979)
Nels R. Olson, 2178 Overlook Ct, Ann Arbor,
MI 48103-2336
Ernest A. Weymuller, Jr, Univ of Washington,
Dept of Oto, BB-1165 Health Sci Bldg, 1959
NE Pacific St, Box 356515, Seattle, WA 981956515
1991
(1964)
2003
(1978)
James L. Parkin, 2390 Bernadine Dr, Salt Lake
City, UT 84109
John R. Williams, The Brittany, Unit 304, 4021
Gulf Shore Blvd N, Naples, FL 33940-3414
1991
(1965)
M. Lee Williams, PO Box 723, Irvington, VA
22480-0723
2006
(1979)
Eiji Yanagisawa, 25 Hickory Rd, Woodbridge,
CT 06525
2000
(1970)
C. Thomas Yarington, Jr, 2136 38th Ave E,
Seattle, WA 98112-2413
2003
(1973)
Anthony J. Yonkers, 508 Ridgewood Dr, Bellevue, NE 68005-4711
2002
(1984)
Victor Passy, 101 The City Dr S, #29E, Orange,
CA 92868-3201
1990
(1963)
Claude Pennington, ENT Ctr of Central Georgia, 800 First St, PO Box 1916, Macon, GA
31202-1916
1985
(1954)
Loring W. Pratt, 37 Lawrence Ave, Fairfield,
ME 04937-1245
1994
(1974)
Robert Priest, 1928 E River Terrace, Minneapolis, MN 55414-3672
1975
(1947)
F. Johnson Putney, 991 Harbortowne Rd,
Charleston, SC 29412
2003
Mona M. Abaza, 7700 E 6th Ave Pkwy, Denver, CO
80230
1983
(1959)
Richard A. Rasmussen, 946 Bellclaire Ave SE,
Grand Rapids, MI 49506-3104
1989
Elliott Abemayor, 12169 Greenock Lane, Los Angeles,
CA 90049-2849
1993
(1969)
Frank N. Ritter, 2675 Englave Dr, Ann Arbor,
MI 48103-2221
2000
Jean Abitbol, 1 Rue Larguilliere, 75016 Paris, France
2008
Lee Akst, Loyola Univ Med Ctr, Dept of Oto, 2160 S
First Ave, Maywood, IL 60153
1968
Bobby R. Alford, Baylor College of Med, Dept of Oto,
2003
(1974)
Robert J. Ruben, Montefiore Med Ctr, Dept of
Oto, 3400 Bainbridge Ave, 3rd Fl, Bronx, NY
Active Members
150
Members
One Baylor Plaza, NA102, Houston, TX 77030-3498
2003
Kenneth W. Altman, Mt Sinai Sch of Med, Dept of Oto,
One Gustave L Levy Place, Box 1189, New York, NY
10029-6500
2003
Milan R. Amin, NYU Voice Ctr, NYU Sch of Med, 550
First Ave, NBV5, East 5, New York, NY 10016
1998
of OSU, 2351 E 22nd St, Cleveland, OH 44115-3111
1996
Orval E. Brown, Southwestern Med Ctr at Dallas, Dept of
Oto, 5323 Harry Hines Blvd, Dallas, TX 75235-9035
1999
J. Dale Browne, Dept of Oto, Wake Forest Univ Sch of
Med, Medical Center Blvd, Winston-Salem, NC 271570001
Vinod K. Anand, 251 Highland Meadow Rd, Flora, MS
39071
1995
Brian B. Burkey, VUMC, Dept of Oto, S-2100 Med Ctr
N, Nashville, TN 37232-2559
2006
Timothy Anderson, 1 Hay Rd, Belmont, MA 02478
2005
1991
Mario Andrea, Av Elias Garcia 57-48, 1000-148 Lisbon,
Portugal
James A. Burns, Dept of Surg, Ctr for Laryngeal Surgery
& Voice Rehab, 208 Village Ave, Dedham, MA 020264231
2000
Donald J. Annino, 47 Valley Rd, Wellesley, MA 02481
2000
1997
Max M. April, 115 Bacon Rd, Old Westbury, NY
11568
Nicholas Busaba, Mass Eye & Ear Infirmary, 243 Charles
St, Boston, MA 02114
1975
1999
Ellis M. Arjmand, Cincinnati Children’s Hosp Med Ctr,
3333 Burnet Ave, ML 2018, Cincinnati, OH 45229
David D. Caldarelli, Dept of Oto, Rush-Presbyterian
Hosp, 1653 W Congress Pkwy, Chicago, IL 60612-3833
1979
Rinaldo Canalis, 457 15th St, Santa Monica, CA 90402
Ricardo Carrau, Ohio State Univ, 456 W 10th Ave, Rm
4024, Columbus, OH 43210
1993
James E. Arnold, 31449 Shaker Blvd, Pepper Pike, OH
44124
2001
1996
Jonathan E. Aviv, 200 West End Ave, Apt 17C, New
York, NY 10023-4801
1997
Paul F. Castellanos, Div of Oto, Univ of Maryland, 16 S
Eutaw St, Ste 500, Baltimore, MD 21201-1619
2006
James Batti, Connecticut Children’s Med Ctr, 282 Washington St, Hartford, CT 06106
2007
Dinesh K. Chhetri, UCLA Sch of Med, Div of Head &
Neck Surg, 62-132 CHS, Los Angeles, CA 90095
1997
Nancy Bauman, Dept of Oto, Univ of Iowa Hosp, 200
Hawkins Dr, Iowa City, IA 52242-1009
2008
Ajay E. Chitkara, 23 Driftwood Lane, East Setauket, New
York, NY 11733
2009
Richard A. Beck, Advanced Oto Services, PA, 3627 University Blvd S, Ste 210, Jacksonville, FL 32216-4211
1997
1989
Stephen P. Becker, Otolaryngology Associates, 233 E
Erie St, Ste 804, Chicago, IL 60611-2906
Sukgi Choi, Children’s Natl Med Ctr, Dept of Oto, 111
Michigan Ave NW, Ste 800, Washington, DC 200102970
1990
2006
Peter Belafsky, UC Davis, 2521 Stockton Blvd, #7200,
Sacramento, CA 95817
Lanny G. Close, Columbia Univ, Dept of Oto, 630 W
168th St, New York, NY 10032
1993
1988
Thomas P. Belson, 1111 Delafield St, Ste 102, Waukesha,
WI 53188-3402
Sharon L. Collins, 6 Timber Shadows Rd, Belleville, IL
62221
1993
1990
Gerald S. Berke, Div of Oto-HNS, UCLA, 10833 Le
Conte Ave, Rm 62-132 CHS, Los Angeles, CA 90095
Stephen F. Conley, 9000 W Wisconsin Ave, PO Box
1997, Milwaukee, WI 53201
1978
1997
Robert G. Berkowitz, Dept of Oto, Royal Children’s
Hosp, Flemington Rd, Parkville, Victoria 3161, Australia
Robin T. Cotton, Children’s Hosp Med Ctr, Dept of Oto,
3333 Burnet Ave, OSB 3110, Cincinnati, OH 45229
1995
Mark S. Courey, UCSF Voice Ctr, 2330 Post St, 5th Fl,
San Francisco, CA 94115-1809
1990
David J. Beste, Children’s Hosp of Wisconsin Office
Bldg, 9000 W Wisconsin Ave, Ste 208, Milwaukee, WI
53226
1991
Dennis M. Crockett, 4650 Sunset Blvd, Los Angeles, CA
90027
1992
1999
Neil Bhattacharyya, Joint Ctr for Oto, 333 Longwood
Ave, 3rd Fl, Boston, MA 02115-5711
James P. Cuyler, 1849 NW Kearney, Ste 300, Portland,
OR 97209-1412
2004
2008
Martin A. Birchall, UCL Ear Institute, Royal Natl
Throat, Nose & Ear Hosp, 330 Gray’s Inn Rd, London,
WC1X 8DA
Seth H. Dailey, Univ of Wisconsin Hosp & Clinics,
K41713 CSC, 600 Highland Ave, Madison, WI 537927375
2006
1990
Jeffrey W. Birns, 5400 Balboa Blvd, Ste 126, Encino, CA
91316-1528
Edward Damrose, 260 Mosher Way, Palo Alto, CA
94304
2000
1988
Andrew Blitzer, NY Ctr for Voice and Swallowing, 425
W 59th St, 10th Fl, New York, NY 10019-1128
David H. Darrow, Eastern Virginia Med Sch, Dept of
Oto, 825 Fairfax Ave, Norfolk VA 23507
1995
R. Kim Davis, 6459 S 640 E, Murray, UT 84107
2003
Joel H. Blumin, Med College of Wisconsin, 9200 W
Wisconsin, Milwaukee, WI 53226
2009
Alessandro de Alarcon, Cincinnati Childrens Hosp,
MLC 2018, 3333 Burnet Ave, Cincinnati, OH 45229
1987
Ronald S. Bogdasarian, Michigan Oto Surg Assoc, 5333
McAuley Dr, Ste 2017, PO Box 994, Ann Arbor, MI
48106-0994
Linda Brodsky, 65 Le Brun Circle, Amherst, NY 14226
Michael Broniatowski, Cleveland Clinic Health Sci Ctrs
2000
Bernard DeBerry, 16300 Sand Canyon, Ste 909, Irvine,
CA 92618
Ziad E. Deeb, Washington Hosp Ctr, Dept of Oto, 110
Irving St NW, Washington, DC 20010
Mark D. Delacure, New York Univ Med Ctr, Dept of Oto,
1993
1998
1999
2003
Members
151
530 First Ave, Ste 7U, Skirball Bldg, New York, NY
10016
2008
Christine Gail Gourin, Med College of Georgia, 1120
16th St, BP 4109, Augusta, GA 30912
2003
Craig S. Derkay, Eastern Virginia Med Sch, Dept of Oto,
825 Fairfax Ave, Norfolk, VA 23507
2010
1998
Daniel E. Deschler, 1347 Massachusetts Ave, Lexington,
MA 02420-3000
Nazaneen Grant, Georgetown Univ Hosp, Dept of Oto,
3800 Reservoir Rd NW, 1 German, Washington, DC
20007
2003
John H. Greinwald, Jr, 9067 Symmes Ridge Lane,
Symmes, OH 45140
1998
Gregory A. Grillone, Dept of Oto, Boston Med Ctr, D616,
88 E Newton St, Boston, MA 02118-2308
1993
Benjamin Gruber, 111 N Wabash, Ste 1603, Chicago, IL
60602-2002
2008
Stacey Halum, Indiana Univ, Dept of Oto-HNS, 702
Barnhill Dr, Ste 860, Indianapolis, IN 46202
1997
Ellen Deutsch, Dept of Ped Oto, Alfred I. duPont Hosp
for Children, 1600 Rockland Rd, POB 269, Wilmington,
DE 19899
2010
Frederik G. Dikkers, Univ Med Ctr Groningen, Univ
of Groningen, PO Box 30.001, 9700 RB Groningen,
Netherlands
1997
Oscar Dias, Dept of Oto, Univ of Lisbon, Av Egas Moniz, Lisbon 1600, Portugal
2000
1998
Donald T. Donovan, Dept of Oto & Communication
Sci, One Baylor Plaza, NA 102, Houston, TX 770303411
David J. Halvorson, 1010 First St N, Ste 301, Alabaster,
AL 35007-8725
1983
Edward Doolin, Dept Ped General & Thoracic Surg,
Children’s Hosp of Philadelphia, 34th & Civic Ctr Blvd,
Philadelphia, PA 19104
Steven D. Handler, Children’s Hosp of Philadelphia,
Richard D. Wood Ctr, 1st Fl, 34th St & Civic Center Blvd,
Philadelphia, PA 19104-4343
1999
Gady Har-El, 193-38 Keno Ave, Hollis, NY 11423
Amelia F. Drake, Univ of North Carolina, Dept of OtoHNS, 170 Manning Dr CB7070, Physicians Office Bldg,
Rm G-116, Chapel Hill, NC 27599-7070
1997
Earl Harley, Dept of Oto, Georgetown Univ Med Ctr,
3800 Reservoir Rd NW, Washington, DC 20007-2196
2004
1991
Michael E. Dunham, 9300 Valley Children’s Place SE18,
Madera, CA 93636
Christopher J. Hartnick, Dept of Oto, Mass Eye & Ear
Infirmary, 243 Charles St, Boston, MA 02114
2003
1986
Roland D. Eavey, Mass Eye & Ear Infirmary, Dept of
Oto, 243 Charles St, Boston, MA 02114-3002
Bruce H. Haughey, Washington Univ Sch of Med, Dept
of Oto, 517 S Euclid Ave, Box 8115, St Louis, MO 63110
1978
1995
David E. Eibling, Eye & Ear Inst, 200 Lothrop St, #500,
Pittsburgh, PA 15213-2548
Gerald B. Healy, Children’s Hosp, Dept of Oto, 300
Longwood Ave, No. 5, Boston, MA 02115-5724
2002
1994
David W. Eisele, 601 N Caroline St, Ste 6210, Baltimore,
MD 21287
Diane G. Heatley, Div of Oto-HNS, Univ of Wisconsin
Hosp, 600 Highland Ave, K4-710, Madison, WI 537927375
2009
Ravindhra G. Elluru, Cincinnati Childrens Hosp, MLC
2018, 3333 Burnet Ave, Cincinnati, OH 45229
2004
Yolanda Heman-Ackah, Philadelphia Voice Ctr, 25 Bala
Ave, Ste 200, Bala Cynwyd, PA 19004
1979
Willard E. Fee, Jr, Stanford Cancer Ctr, 875 Blake Wilburn Dr, Rm CC-2227, Stanford, CA 94306-5826
1991
Robert A. Hendrix, 105 Shannon Ct, Rocky Mount, NC
27804
2000
James W. Forsen, Jr, 621 S New Ballas Rd, Ste 622A, St
Louis, MO 63141
1980
Arthur S. Hengerer, Dept of Oto, 601 Elmwood Ave,
Box 629, Rochester, NY 14642-0629
1985
Marvin P. Fried, Montefiore Med Ctr, Dept of Oto, 3400
Bainbridge Ave, 3rd Fl, Bronx, NY 10467-2490
1997
Garrett D. Herzon, Cedars-Sinai Med Ctr, Ste 440E, 8631
W Third St, Los Angeles, CA 90048
1985
Ellen M. Friedman, 2521 Reba Dr, Houston, TX 77019
1997
Raymond L. Hilsinger, Jr, HNS Dept, 280 W MacArthur
Blvd, Oakland, CA 94611-5642
1990
Michael Friedman, 30 N Michigan Ave, Ste 1107, Chicago, IL 60602
2003
Michael L. Hinni, Mayo Clinic Scottsdale, 5777 E Mayo
Blvd, Phoenix, AZ 85054
1999
C. Gaelyn Garrett, 7302 Med Ctr E, South Tower, Nashville, TN 37212
2002
Shigeru Hirano, 51-2 Hirano-Toriimae-Cho #405, Kitaku, Kyoto 603-8321, Japan
1986
Kenneth A. Geller, c/o CHLAMS #78, 4650 Sunset Blvd,
Los Angeles, CA 90027
1999
Henry T. Hoffman, Dept of Oto, Univ of Iowa Hosp, 200
Hawkins Dr, Iowa City, IA 52242-1009
2002
Eric M. Genden, Dept of Oto, Mt Sinai Med Ctr, 1
Gustave L. Levy Pl, New York, NY 10029-6574
1978
Lauren D. Holinger, Dept of Oto, Children’s Memorial
Hosp, 2300 Children’s Plaza, Box 25, Chicago, IL 60614
2003
Mark E. Gerber, Ped Oto-HNS, North Shore Univ Health
System, 501 Skokie Blvd, Northbrook, IL 60062
1993
Andrew J. Hotaling, 18 S Clay St, Hinsdale, IL 605213235
1984
Carol R. Gerson, 1031 Ashland Ave, River Forest, IL
60305
1991
Andrew F. Inglis, Children’s Hosp & Regional Med Ctr,
PO Box 5371 1 6E-1, Seattle, WA 98105-0371
1992
W. Jarrard Goodwin, Jr, Univ of Miami, No. 4037, 1475
NW 12th Ave, Miami, FL 33136-1002
1997
2009
Joshua A. Gottschall, Kaiser Permanente Med Ctr, ORLHNS, 2800 W MacArthur Blvd, Oakland, CA 94611
Ian Jacobs, Dept of Ped Oto, Children’s Hosp of Philadelphia, 34th St & Civic Center Blvd, 1st Fl Wood, Philadelphia, PA 19104
1976
Bruce W. Jafek, Univ of Colorado, Dept of Oto B-205,
1995
2003
152
Members
4200 E Ninth Ave, Denver, CO 80262
1989
2009
Scharukh Jalisi, Boston Univ Med Ctr, ORL-HNS, 820
Harrison Ave, Ste FGH4, Boston, MA 02118
Howard L. Levine, Cleveland Nasal Sinus & Sleep Ctr,
5555 Transportation Blvd, Cleveland, OH 44125-5325
1990
2005
Michael M. Johns, 550 Peachtree St, Ste 400, Atlanta,
GA 30308
Paul A. Levine, UVA, Oto-HNS, Box 800713, Charlottesville, VA 22908-0713
1989
Rodney P. Lusk, 555 N 30th St, Omaha, NE 68131
1985
Jonas T. Johnson, Eye & Ear Inst, Dept of Oto, 200
Lothrop St, Ste 500, UPMC, Pittsburgh, PA 15213-2546
2008
2000
Paul J. Jones, Rush Univ Med Ctr, Dept of Ped Oto, 1611
W Harrison, Ste 550, Chicago, IL 60612-3833
David L. Mandell, Ctr for Ped ENT, Ste B-900, Bethesda
Health City, 10301 Hagen Ranch Rd, Boynton Beach, FL
33437
2009
1991
Raleigh O. Jones, Jr, Univ of Kentucky Med Ctr, Div of
Oto, 800 Rose St, Rm C-236, Lexington, KY 40536-0084
Nicolas Maragos, Mayo Clinic, 200 First St SW, Rochester, MN 55905
1995
2004
David E. Karas, 14 Willard Ave, Madison, CT 06443
1999
Jan L. Kasperbauer, Mayo Clinic, Dept of Oto, 200 First
St SW, Rochester, MN 55905-0002
Lynette J. Mark, Johns Hopkins UMC, Div of Anesthesia
for Oto-HNS, 600 N Wolfe St, Tower 711, Baltimore,
MD 21287-8711
2003
1973
Burns W. Kay, One W Hellman Ave, No. 8, Alhambra,
CA 91803
Nicole C. Maronian, Univ of Cleveland, 11100 Euclid
Ave, LKS 5045, Cleveland, OH 44106-5045
2005
1997
William M. Keane, 925 Chestnut St, 6th Fl, Philadelphia,
PA 19107
Steffen Maune, Univ of Kiel, Dept of Oto, Arnold-Heller-Str 14, 24105 Kiel, Germany
1984
1992
Donald B. Kearns, 3030 Children’s Way, Ste 402, San
Diego, CA 92123-4228
Thomas V. McCaffrey, Univ of Southern Florida, 12902
Magnolia Dr, Ste 3057, Tampa, FL 33612-9416
2008
1993
James H. Kelly, 6701 N Charles St, GBMC Rm 4202,
Baltimore, MD 21204
Timothy M. McCulloch, Univ of Wisconsin Hosp & Clinics, K4/719 Clinical Sci Ctr, 600 Highland Ave, Madison,
WI 53792-7375
1998
David W. Kennedy, Univ of Pennsylvania Med Ctr, Dept
of Oto, 3400 Spruce St, Ravdin 5, Philadelphia, PA
19104-4264
1982
John C. McDougall, Mayo Clinic, 200 SW First St,
Rochester, MN 55905
1984
Trevor J. I. McGill, Children’s Hosp, Dept of Oto, Fegan
9, 300 Longwood Ave, Boston, MA 02115-5724
1998
Kemp H. Kernstine, City of Hope Natl Med Ctr, 1500 E
Duarte Rd, Warshaw MOB, Duarte, CA 91010
1998
1998
Joseph E. Kerschner, Dept of Ped Oto, Children’s Hosp
of Wisconsin, 9200 W Wisconsin Ave, Milwaukee, WI
53226-4810
William Frederick McGuirt, Jr, 4308 Allstair Rd, Winston-Salem, NC 27104
2000
1984
Charles P. Kimmelman, 985 Fifth Ave, 7A, New York,
NY 10021
J. Scott McMurray, Div of Oto-HNS, Univ of Wisconsin
Hosp, 600 Highland Ave, K4-710, Madison, WI 537927375
2003
2008
Adam M. Klein, Emory Voice Ctr, 550 Peachtree St,
MOT, Ste 9-4400, Atlanta, GA 30308
2008
1992
Peter J. Koltai, Stanford Univ Sch of Med, Div of
Ped Oto, 801 Welch Rd, Stanford, CA 94305-5739
2000
1988
Arnold Komisar, 1421 Third Ave, 4th Fl, New York, NY
10028
1990
1990
Charles F. Koopmann, 2514 Hawthorne Rd, Ann Arbor,
MI 48104
2005
1989
Jamie Koufman, 121 Bank St, Apt K, New York, NY
10014
1996
Dennis H. Kraus, Memorial Sloan-Kettering Cancer Ctr,
1275 York Ave, Box 285, New York, NY 10021-0032
1984
1989
Yosef P. Krespi, NYHNI, 110 E 59th St, Ste 8C, New
York, NY 10022
1997
2010
Priya D. Krishna, UPMC Mercy Hosp, Dept of Oto, Ste
11500, Bldg B, 1400 Locust St, Pittsburgh, PA 15219
1993
Frederick A. Kuhn, Med College of Georgia, Div of
ENT, 4750 Waters Ave, BP 4117, Ste 112, Savannah, GA
31404-6267
2008
Denis C. LaFreniere, Univ of Connecticut Health Ctr,
Div of Oto-HNS, MC-6228, 263 Farmington Ave,
Farmington, CT 06032-6228
1988
William Lawson, Dept of Oto, Mt Sinai Med Ctr, Five E
98th St, 8th Fl, Box 1191, New York, NY 10029
Albert L. Merati, Univ of Washington Sch of Med, 1959
NE Pacific Dr, Seattle, WA 98195-6161
Tanya K. Meyer, 10 E Lee St, #1101, Baltimore, MD
21202
Henry A. Milczuk, Dept of Oto-HNS, Oregon Health Sci
Univ, PV-01, 3181 SW Sam Jackson Park Rd, Portland,
OR 97201-3011
Robert P. Miller, 8780 Golf Rd, Ste 200, Niles, IL 607145611
Natasha Mirza, MD, Univ of Pennsylvania Med Ctr,
Voice & Swallowing Ctr, 5 Ravdin, 3400 Spruce St,
Philadelphia, PA 19104
Rose M. Mohr, Dept of Oto, Campus Med Arts Bldg #2,
Ste 230, 29101 Health Campus Dr, Westlake, OH 44145
Anthony J. Mortelliti, Dept of Oto, SUNY Upstate Med
Univ, 750 E Adams St, Syracuse, NY 13210
Melissa Mortensen, Univ of Virginia Health System,
ORL-HNS, PO Box 800713, Charlottesville, VA 229080713
Harlan R. Muntz, Primary Children’s Med Ctr, 100 N
Mario Capecchi Dr, #4500, Salt Lake City, UT 84113
Charles M. Myer III, Children’s Hosp Med Ctr, Dept of
Ped Oto, 3333 Burnet Ave, ML 2018, Cincinnati, OH
45229-3026
James L. Netterville, VUMC, Dept of Oto, 7209 Med Ctr
E, South Tower, 1215 21st Ave S, Nashville, TN 372325555
2009
1991
1994
1993
Members
2009
J. Pieter Noordzij, Boston Med Ctr, ORL-HNS, 715
Albany St, Boston, MA 02118
1995
Laurie A. Ohlms, Children’s Hosp, Dept of Oto, 300
Longwood Ave, Boston, MA 02115
2006
Bert O’Malley, Univ of Pennsylvania, 3400 Spruce St, 5
Ravdin, Philadelphia, PA 19104
2007
153
1986
Mark A. Richardson, Dept of Oto, Oregon Health Sci
Univ, PV-01, 3181 SW Sam Jackson Park Rd, Portland,
OR 97239
2009
Gresham T. Richter, Univ of Arkansas for Med Sci, Ped
Oto, Arkansas Children’s Hosp, 4301 W Markham, Slot
543, Little Rock, AR 72205
Laura Orvidas, Mayo Clinic, Dept of Oto, 200 First St
SW, Rochester, MN 55905
1994
William J. Richtsmeier, Bassett Healthcare, One Atwell
Rd, Cooperstown, NY 13326-1301
1984
Robert H. Ossoff, VUMC, S-2100 Med Ctr N, Nashville,
TN 37232-2559
1994
Marion B. Ridley, 3203 Bayshore Blvd #1802, Tampa,
FL 33629
2001
Randal C. Paniello, Washington Univ Med Ctr, Dept of
Oto, 660 Euclid Ave, Box 8115, St Louis, MO 631101007
1998
Frank L. Rimell, 420 Delaware St SE, MMC 396, Minneapolis, MN 55455-0374
1976
2000
Albert H. Park, Primary Children’s Med Ctr, Div of Oto,
100 N Mario Capecchi Dr, #4500, Salt Lake City, UT
84113
Eugene Rontal, 28300 Orchard Lake Rd, No. 100, Farmington Hills, MI 48334-3704
1981
Michael Rontal, 28300 Orchard Lake Rd, No. 100, Farmington Hills, MI 48334-3704
1990
Steven M. Parnes, 389 Hanley Rd, Nassau, NY 12123
2003
1998
Thomas R. Pasic, Univ of Wisconsin Hosp, 600 Highland
Ave, K4-712, Madison, WI 53792-7375
Kristina W. Rosbe, Univ of California–San Francisco,
Dept of Oto, Ste A730, San Francisco, CA 94143-0342
1998
1987
Mark S. Persky, Beth Israel Med Ctr, 10 Union Sq E, Ste
4J, New York, NY 10003
Clark A. Rosen, Eye & Ear Inst, 200 Lothrop St, Ste 500,
Pittsburgh, PA 15213
1999
1994
Glenn Edison Peters, Univ of Alabama at Birmingham,
BDR 563, 1530 3rd Ave, Birmingham, AL 35294-0012
Richard M. Rosenfeld, Dept of Oto, 339 Hicks St, Brooklyn, NY 11201
2004
1984
Harold C. Pillsbury III, 103 Turtleback Crossing Dr,
Chapel Hill, NC 27516
Douglas A. Ross, St Vincent, Dept of Surg, 2800 Main
St, Bridgeport, CT 06606-4201
1998
1991
Robert L. Pincus, 36A E 36th St, Ste 200, New York, NY
10016-3463
Michael Rothschild, MD, Park Ave ENT, 1175 Park Ave,
Ste 1A, New York, NY 10128
2005
2009
Michael Pitman, New York Eye & Ear Infirmary, 310 E
14 St, New York, NY 10003
John S. Rubin, 15 Smith Close, London SE 165 PB,
England
2004
1999
William M. Portnoy, 160 W 18th St, New York, NY
10011-5403
Michael J. Rutter, Cincinnati Children’s Hosp, Dept of
Ped Oto, 3333 Burnet Ave, ML 2018, Cincinnati, OH
45229
1998
Gregory Postma, Med College of Georgia, Dept of Oto,
1120 15th St, BP-4109, Augusta, GA 30912
2003
Alain Sabri, American Univ of Beirut Med Ctr, Dept of
Oto-HNS, PO Box 113-6044, Beirut, Lebanon
1997
William Potsic, Div of Oto, Children’s Hosp of Philadelphia, 34th St & Civic Center Blvd, Richard D. Wood
Bldg, Philadelphia, PA 19104-4303
1989
Clarence T. Sasaki, Yale Univ Sch of Med, Section of
Oto, 333 Cedar St, PO Box 208041, New Haven, CT
06520-8041
1992
Seth M. Pransky, 3030 Children’s Way, Ste 402, San
Diego, CA 92123-4228
1997
Robert Sataloff, 1721 Pine St, Philadelphia, PA 191036701
2002
Reza Rahbar, Dept of Oto, Children’s Hosp, 300 Longwood Ave, Boston, MA 02115
2000
Kiminori Sato, 8-18-2-chome Kanaike-Machi, Oita-Shi
870-0026, Japan
2001
Elie E. Rebeiz, New England Med Ctr, 302 Marlborough
St, Boston, MA 02116-1607
1996
Richard L. Scher, Div of Oto, Duke Univ Med Ctr, Box
3805, Durham, NC 27710
2009
Catherine J. Rees, Wake Forest Univ Health Sci, Med
Ctr Blvd, Winston, NC 27157
1998
Scott R. Schoem, 245 Westmont St, West Hartford, CT
06117
2003
Mark Reichelderfer, Univ of Wisconsin Hosp, 600 Highland Ave, H6/516 CSC, Madison, WI 53792-5124
2005
John M. Schweinfurth, Univ of Mississippi, Dept of Oto,
2500 N State St, Jackson, MS 39216-4505
1980
Timothy J. Reichert, 621 S New Ballas Rd, Ste 383A, St
Louis, MO 63141-8232
1983
1986
James S. Reilly, Alfred I. duPont Hosp for Children,
1600 Rockland Rd, Wilmington, DE 19899
1996
Anthony J. Reino, 175 Memorial Hwy, Ste 1-2, New
Rochelle, NY 10801
1997
Marc Remacle, Service ORL, UCL Mont-Godinne, Av
Therasse 1, Yvoir 5530, Belgium
1980
Dale H. Rice, USC Health Consultation Ctr, 1510 San
Pablo St, Ste 201, Los Angeles, CA 90033-4586
Roy B. Sessions, Beth Israel Med Ctr, 10 Union Sq E, Ste
45, New York, NY 10003-3314
Michael Setzen, 600 Northern Blvd, Ste 310, Great Neck,
NY 11021-5200
Udayan K. Shah, Alfred I duPont Hosp for Children, Div
of Oto, 1600 Rockland Rd, Wilmington, DE 19803
Jo Shapiro, Div of Oto, Brigham & Women’s Hosp, 45
Francis St, Boston, MA 02115-5711
Nina Shapiro, Div of HNS, UCLA Med Ctr, 10833 Le
Conte Ave, Rm 62-158 CHS, Los Angeles, CA 900953075
1988
1998
1998
1998
154
Members
1984
Stanley M. Shapshay, University ENT, 35 Haskett Blvd,
Albany, NY 12208
of Med, 601 Caroline St, 6th Fl, Baltimore, MD 212870910
2006
Anat Shatz, Shaare Zedek Med Ctr, Dept of Oto, 78
Hagalil St, Ganey Tikva 55900, Israel
2010
V. Jean Verheyden, Central Oregon ENT LLC, 2450 NE
Mary Rose Place, Ste 120, Bend, OR 97701
2000
Gary Y. Shaw, 11911 W 139 Terr, Overland Park, KS
66221
2000
David L. Walner, 1680 Cloverdale Ave, Highland Park,
IL 60035
2006
Akihiro Shiotani, 5-3-1 Nogata Nakano, Tokyo 1650027,
Japan
1980
Ko-Pen Wang, Harbor Hosp Ctr, 3001 S Hanover St,
Baltimore, MD 21225
1993
William W. Shockley, Div of Oto, Univ of North Carolina, CB 7070, 610 Burnett-Womack Bldg, Chapel Hill,
NC 27599-7070
1995
Robert F. Ward, Weill Cornell Med College, Dept of Oto,
1305 York Ave, 5th Floor, New York, NY 10021
1998
2000
Sally R. Shott, MD, Children’s Hosp Med Ctr, 3333
Burnet Ave, MCL 2018, Cincinnati, OH 45229-3039
Mark K. Wax, Dept of Oto, 3181 SW Sam Jackson Park
Rd, PV-01, Portland, OR 97201-3098
2004
2010
Jeffrey P. Simons, Langone Med Ctr, Ped Oto-NYU, 550
First Ave, New York, NY 10016
Julie L. Wei, Univ of Kansas Med Ctr, Dept of Oto,
Mailstop 3010, 3901 Rainbow Blvd, Kansas City, KS
66160
2000
C. Blake Simpson, Univ of Texas Health Sci Ctr, 7703
Floyd Curl Dr, MSC 777, San Antonio, TX 78229-3900
1996
1984
George T. Simpson II, 3495 Bailey Ave 503-C, Buffalo,
NY 14215
Gregory S. Weinstein, Dept of Oto, Univ of Pennsylvania
Med Ctr, 5 Ravdin, 3400 Spruce St, Philadelphia, PA
19104-4219
1984
2010
John T. Sinacori, 600 Gresham Dr, Ste 1100 RP, Norfolk,
VA 23507
Robert A. Weisman, 5215 Cannito Vista Lujo, San Diego,
CA 92130
1993
2003
Marshall E. Smith, Univ of Utah Med Ctr, Div of Oto, 50
N Mario Capecchi Dr, Rm 3C120 SOM, Salt Lake City,
UT 84132
Mark C. Weissler, 100 Faison Rd, Chapel Hill, NC
27517
1991
Barry L. Wenig, Evanston Northwestern Hosp, Div of
Oto, 1000 Central, Ste 610, Evanston, IL 60201
1980
Raymond O. Smith, 4200 W Memorial Rd, No. 606,
Oklahoma City, OK 73120-8305
1995
Jay A. Werkhaven, VUMC, Dept of Oto, 7209 Med Ctr
E, Nashville, TN 37232-2559
1990
Richard J. H. Smith, Dept of Oto, Univ of Iowa Hosp, 200
Hawkins Dr, Iowa City, IA 52242-1078
1999
2004
Ahmed Soliman, Temple Univ Sch of Med, Dept of
Oto-HNS, 3400 N Broad St, Kresge 102, Philadelphia,
PA 19140
Ralph Wetmore, Children’s Hosp of Philadelphia, Dept
of Ped Oto, 34th St & Civic Center Blvd, Philadelphia,
PA 19104
1997
Brian Wiatrak, Ped ENT Associates, 1940 Elmer J Bissell
Rd, Birmingham, AL 35243
2007
Robert Stachler, 527 N Cranbrook Dr, Bloomfield Hills,
MI 48301-2402
2000
J. Paul Willging, Children’s Hosp Med Ctr, 3333 Burnet
Ave, MCL 2018, Cincinnati, OH 45229
1987
James A. Stankiewicz, Dept of Oto, Loyola Univ Med
Ctr, 2160 S First Ave, Maywood, IL 60153-3304
1997
Daniel Wohl, 4114 Sunbeam Rd, Ste 403, Jacksonville,
FL 32257
1981
Marshall Strome, 450 E 83rd St, Apt 23C, New York,
NY 10020-6293
1993
Peak Woo, 143 Hudson Ave, Tenafly, NJ 07670
2001
W. Edward Wood, 22 Lindsey Ave, Danville, PA 17821
1978
Fred J. Stucker, Louisiana State Univ Med Ctr, Dept of
Oto-HNS, 1501 Kings Hwy, Shreveport, LA 71103
2000
2004
Lucian Sulica, Weill Cornell Med College, Dept of Oto,
1305 York Ave, 5th Floor, New York, NY 10021
B. Tucker Woodson, Med College of Wisconsin, Clinic
at Froedtert, 9200 W Wisconsin Ave, Milwaukee, WI
53226
2002
2007
Dana Suskind, Univ of Chicago, Oto-HNS, 5841 S Maryland Ave, M/C 1035, Chicago, IL 50537
Gayle W. Woodson, 1317 Wiggins Ave, Springfield, IL
62704
1999
2008
Thomas G. Takoudes, 46 Prince St, Ste 601, New Haven,
CT 06519
Audie L. Woolley, Ped ENT Associates, 1940 Elmer J
Bissell Rd, Birmingham, AL 35243
1997
2008
Ichiro Tateya, Dept of Oto-HNS, Graduate Sch of Medicine, Kyoto Univ, Sakyo-ku, Kyoto 606-8507, Japan
Ken Yanagisawa, Univ Towers, 98 York St, New Haven,
CT 06511-5602
2010
2000
David J. Terris, Georgia Health Sci Univ, Dept of Oto,
1120 Fifteen St, BP-4109, Augusta, GA 30912-4060
Nwanmegha Young, Yale Univ, Dept of Oto, 333 Cedar
St, PO Box 208041, New Haven, CT 06520-8041
2009
2000
Dana M. Thompson, 1402 Autumn Sage Courts W,
Rochester, MN 55902
Craig H. Zalvan, Institute for Voice & Swallowing Disorders, 777 N Broadway, Ste 303, Sleepy Hollow, NY
10591
1985
Jerome W. Thompson, UT Health Sci Ctr, Dept of Oto,
956 Court Ave, Rm B226, Memphis, TN 38163
1997
George Zalzal, Children’s Natl Med Ctr, Dept of Oto,
111 Michigan Ave NW, Washington, DC 20010
1976
Robert J. Toohill, Med College of Wisconsin, Clinic at
Froedtert, 9200 W Wisconsin Ave, Milwaukee, WI 53226
1991
Steven M. Zeitels, 100 Bellevue St, Boston, MA 024581921
1996
David Tunkel, Dept of Oto, Johns Hopkins Univ Sch
2006
Karen Zur, Children’s Hosp of Philadelphia, Richard D.
Members
Wood Ctr, 1st Floor, 34th St & Civic Ctr Blvd, Philadelphia, PA 19104-4399
1992
David A. Zwillenberg, 266 Linden Lane, Merion, PA
19066
Associate Members
155
seau, 26 Ave du Dr Arnold Netter, Paris Cedex 75012,
France
2008
Dana M. Hartl, Institut Gustave Roussy, Oto-HNS, 39,
rue Camille Desmoulins, 94805 Villejuif, France
1982
Minoru Hirano, 242-5 Nishimachi, Kurume-shi 830, Japan
2008
Lynn Acton, Yale Univ, Communication Disord Ctr, 333
Cedar St, New Haven, CT 06520
1995
Yasuo Hisa, Dept of Oto, Kyoto Prefectural Univ of Med,
Kawaramachi-Hirokoji, Kyoto 602-8566, Japan
1984
Jerome C. Goldstein, 4119 Manchester Lake Dr, Lake
Worth, FL 33467-8175
2000
Katsuhide Inagi, Kitasato Inst Med Ctr, 6-100 Arai, Kitamoto, Saitama, Japan
1998
Andrew Herlich, Temple Univ, Dept of Anesthesiol, 3rd
Fl, Broad & Ontario Sts, Philadelphia, PA 19104
1973
S. N. Jain, A1 F2 Poonam Apts, Abid Shopping Ctr,
Chiraz Ali Lane, Hyderabad 500001 India
2009
Nikki Johnston, Med College of Wisconsin, Otol &
Commun Sci, Div of Research, Froedtert West, 9200 W
Wisconsin Ave, Milwaukee, WI 53226
1976
Otto Jepsen, Univ Dept of Oto, Arhus Kommunehospital, 8000 Arhus, Copenhagen, Denmark
2008
Steven B. Leder, Yale Univ Sch of Med, Dept of SurgOto, PO Box 208041, New Haven, CT 06520-8041
2005
Benjamin Y. Kim, Yonsei Univ College of Med, Dept
of ENT, CPO Box 8044, Seoul, Korea
2008
Heather Lisitano, Yale Univ, Oto, 800 Howard Ave,
YPB 4th Floor, New Haven, CT 06520-8041
1994
Hisayoshi Kojima, Dept of Oto, Kyoto Univ Fac of Med,
Syogoinn Kawahara-cho 54, Sakyo-ku, Kyoto 606-8507,
Japan
2005
Thomas Murry, Columbia Univ, Dept of Oto, Harkness
8-812, New York, NY 10032
1979
Carl-Eric Lindholm, Dept of Oto, Orebro Med Ctr Hosp,
S-701 85 Orebro, Sweden
2009
Diana Marie Orbelo, Mayo Clinic, 200 First St SW,
Rochester, MN 55905
2004
2001
JoAnne Robbins, William S. Middleton VA Hosp,
GRECC (11G), 2500 Overlook Terrace, Madison, WI
53705
Burkard M. Lippert, SLK-Kliniken Heilbronn GmbH,
Dept of ORL, Am Gesundbrunnen 20-26, 74078 Heilbronn, Germany
1980
Salvador Magaro, R Pena 1279-10B, Buenos Aires 1021,
Argentina
Libby J. Smith, Eye & Ear Institute, 203 Lothrup St, Ste
519, Philadelphia, PA 15123
2002
Hans F. Mahieu, VU Med Ctr, Dept of Oto, POB 7057,
1007 MB Amsterdam, Netherlands
1992
Wolf J. Mann, Dept of Oto, Langenbeckstr, Mainz 55101,
Germany
2008
Corresponding Members
1974
Bruce N. Benjamin, 19 Prince Rd, Killara, NSW 2071,
Australia
1987
1993
Daniel F. Brasnu, HEGP, Dept of Oto, 20 rue Leblanc,
Paris 75015, France
Juan Antonio Mazzei, AV Las Heras 2321, 2nd Fl,
C1425ASA C.A. Buenos Aires, Argentina
1991
1991
G. Patrick Bridger, 21 Kitchener Pl, Ste 1, Bankstown,
NSW 2200, Australia
Randall P. Morton, Green Lane Hosp, ORL Dept, Green
Lane West, Auckland 3, New Zealand
1991
2001
Harvey L. Coates, 208 Hampden Rd, Nedlands, Perth,
WA 6009, Australia
Yasushi Murakami, 4-2 Goryoshita, Ryoanji, U-Kyo-Ku,
Kyoto 616, Japan
2004
2006
Jacob Cohen, 119 Shderot Rothschild St, 64271 TelAviv, Israel
Tadashi Nakashima, Kurume Univ Sch of Med, Dept
of Oto-HNS, 67 Asahi-machi, Kurume 830-0011, Japan
1946
Emiro E. Delima, Rua Alvaro Alvin 31 130, Rio de Janeiro, Brazil
1997
Michael Nash, Dept of Oto, Soroka Univ Med Ctr, Box
151, Beer Sheva 84101, Israel
2004
Ari DeRowe, Tel-Aviv Med Ctr, Dana Children’s Hosp,
6 Weitzman St, Tel-Aviv, Israel
1976
Arnold M. Noyek, Mt Sinai Hosp, 600 University, Rm
401, Toronto, Ontario M5G 1X5, Canada
1965
J. M. DuBois de Montreynaud, Centre Hosp Univ, 45
Rue Cognacq-Jay, Reims Cedex 51090, France
2002
Koichi Omori, Dept of Oto-HNS, Grad Sch of Med,
Kyoto Univ, 54 Kawahara-cho, Shogoin, Kyoto 6068507, Japan
2002
Hans E. Eckel, HNO Abteilung, Dept ORL, Landeskrankenhaus Klagenfurt, St Veiter Str 47, A-9020 Klagenfurt,
Austria
1987
1988
Alfio Ferlito, Dept of Oto-HNS, Univ of Udine, Policlinico
Universitario, Piazzale Solavia Maria della Misericordia,
33100 Udine, Italy
Tadesz M. Orlowski, Dept of Thoracic Surg, 105 Grabiszynska Str, 53-439, Wroclaw, Poland
Alexei Ovchinnikov, Surgery Clinik N1, First Moscow
Medical Institute, Dovator Str 15, Moscow, Russia
Panagiotis E. Pantazepoulos, 54 Homiru St, Athens 135,
Greece
Kishore Prasad, 1st Floor Nethravathi Bldg, Balmatta, Mangalore, Karnataka State, India, 575001
Alessandra Rinaldo, Dept of Oto-HNS, Univ of Udine,
Policlinico Città di Udine, Viale Venezia 410, Udine
33100, Italy
1980
2003
2001
Rolando Fonseca, La Rioja 3920, La Lucila, Buenos
Aires 1636, Argentina
Gerhard Friedrich, Auenbruggerplatz 26-28, A-8036
Graz, Austria
E. Noel Garabedian, Service ORL, Hosp Armand Trous-
1984
1966
2004
2000
156
1974
2005
2002
1991
2004
2005
2000
Members
Marcel Savary, Rue Neuve 10, Yverdon 1400, Switzerland
Christian Sittel, Oto-HNS, Klinkum Stuttgart-Katharinenhospital, Kriegsbergstraße 60, 70174 Stuttgart,
Germany
Conrad F. Smit, Vrije Univ Med Ctr, POB 7057, 1007
MB Amsterdam, Netherlands
Gordon B. Snow, VUMC, Dept of Oto, De Boelelaan
1117, 1081 Amsterdam, 1081-HV, Netherlands
Georg Mathias Sprinzl, Univ of Innsbruck, HNO Klinik
Anichstr, Dept of Oto, 356020 Innsbruck, Austria
Wolfgang Steiner, Univ of Goettingen, ENT Dept, RobertKoch-Str 40, Goettingen 37075, Germany
Juan Manuel Tato, Lago Hess 13 110 110KM 13 1, Av
Bustillo 8400 San Carlos, Rio Negro 8400, Argentina
2002
Jean M. Triglia, Hôpital d’Enfants CHU La Timone,
Dept of Pediatric Oto, 13385 Marseille Cedex 5, France
2004
Hirohito Umeno, Kurume Univ, Dept of Oto-HNS, 67
Asahi-machi, Kurume 830-0011, Japan
1991
Toshiyuki Uno, Kyoto Prefectural Univ, Kawaramachi
Hirokoji, Kamikyo-ku, Kyoto 602-8566, Japan
1993
Jos J. M. van Overbeek, Kosterijweg 9, 9761 GD Eelde,
Netherlands
2003
Jochen Werner, Dept of Oto, Univ of Marburg, Deutschhausstr 3, 35037 Marburg, Germany
2008
Jeong-Soo Woo, 4 Maplecrest Lane, Hamden, CT
06514
OFFICERS 1917-2010
1917
1918
1919
1920
1921
1922
1923
1924
1925
1926
1927
1928
1929
1930
1931
1932
1933
1934
1935
1936
1937
1938
1939
1940
1941
1942
1943
1944
1945
1946
1947
1948
Chevalier Jackson, MD
Hubert Arrowsmith, MD
John W. Murphy, MD
Henry L. Lynah, MD
Harris P. Mosher, MD
Samuel Iglauer, MD
Robert C. Lynch, MD
Ellen J. Patterson, MD
William B. Chamberlin, MD
D. Crosby Greene, MD
Sidney Yankauer, MD
Charles J. Imperatori, MD
Thomas E. Carmody, MD
Henry B. Orton, MD
Louis H. Clerf, MD
Richmond McKinney, MD
Waitman F. Zinn, MD
Henry Hall Forbes, MD
H. Marshall Taylor, MD
Joseph C. Beck, MD
Gordan Berry, MD
John Kernan, MD
Lyman Richards, MD
Gabriel Tucker, MD
W. Likely Simpson, MD
Robert L. Moorehead, MD
Robert L. Moorehead, MD
Carlos E. Pitkin, MD
Carlos E. Pitkin, MD
Robert M. Lukens, MD
Millard F. Arbuckle, MD
Paul H. Holinger, MD
1949
1950
1951
1952
1953
1954
1955
1956
1957
1958
1959
1960
1961
1962
1963
1964
1965
1966
1967
1968
1969
1970
1971
1972
1973
1974
1975
1976
1977
1978
1979
PRESIDENTS
LeRoy A. Schall, MD
Chevalier L. Jackson, MD
Herman J. Moersch, MD
Fred W. Dixon, MD
Edwin N. Broyles, MD
Clyde A. Heatly, MD
Daniel S. Cunning, MD
Clarence W. Engler, MD
Walter B. Hoover, MD
Francis W. Davison, MD
Verling K. Hart, MD
F. Johnson Putney, MD
Alden H. Miller, MD
Joseph P. Atkins, MD
Stanton A. Friedberg, MD
Charles M. Norris, MD
Daniel C. Baker, Jr, MD
Blair Fearon, MD
Francis E. LeJeune, Jr, MD
Charles F. Ferguson, MD
Arthur M. Olsen, MD
Richard W. Hanckel, MD
John R. Ausband, MD
John S. Knight, MD
Gabriel F. Tucker, Jr, MD
Howard A. Anderson, MD
Walter H. Maloney, MD
Seymour R. Cohen, MD
Paul H. Ward, MD
James B. Snow, Jr, MD
Joyce A. Schild, MD
1980
1981
1982
1983
1984
1985
1986
1987
1988
1989
1990
1991
1992
1993
1994
1995
1996
1997
1998
1999
2000
2001
2002
2003
2004
2005
2006
2007
2008
2009
2010
Loring W. Pratt, MD
M. Stuart Strong, MD
Bernard R. Marsh, MD
John A. Tucker, MD
Frank N. Ritter, MD
William R. Hudson, MD
David R. Sanderson, MD
C. Thomas Yarington, Jr, MD
Robert W. Cantrell, MD
H. Bryan Neel III, MD, PhD
Gerald B. Healy, MD
Charles W. Cummings, MD
Lauren D. Holinger, MD
Haskins K. Kashima, MD
Eiji Yanagisawa, MD
Robert H. Ossoff, DMD, MD
Stanley M. Shapshay, MD
Rodney P. Lusk, MD
W. Frederick McGuirt, MD
Paul A. Levine, MD
Ellen M. Friedman, MD
Robin T. Cotton, MD
Peak Woo, MD
Charles N. Ford, MD
Steven M. Zeitels, MD
Jonathan E. Aviv, MD
Gady Har-El, MD
Clarence T. Sasaki, MD
Jamie A. Koufman, MD
Andrew Blitzer, MD, DDS
Michael A. Rothschild, MD
1917
1918
1919
1920
1921
1922
1923
1924
1925
1926
1927
1928
1929
1930
1931
1932
1933
1934
1935
1936
1937
1938
1939
1940
1941
1942
1943
1944
1945
1946
1947
1948
Hubert Arrowsmith, MD
John W. Murphy, MD
Harris P. Mosher, MD
Robert C. Lynch, MD
Robert C. Lynch, MD
Charles J. Imperatori, MD
Fielding O. Lewis, MD
Thomas E. Carmody, MD
D. Crosby Greene, MD
Sidney Yankauer, MD
Henry B. Orton, MD
W. Likely Simpson, MD
H. Marshall Taylor, MD
Richmond McKinney, MD
Edwin McGinnis, MD
D. Campbell Smyth, MD
LeRoy A. Schall, MD
H. Marshall Taylor, MD
Gabriel Tucker, MD
John Kernan, MD
William F. Molt, MD
Edward A. Looper, MD
Dean M. Lierle, MD
Carlos E. Pitkin, MD
Robert L. Moorehead, MD
Fletcher D. Woodward, MD
Fletcher D. Woodward, MD
Kenneth A. Phelps, MD
Kenneth A. Phelps, MD
Murdock Equen, MD
Edwin N. Broyles, MD
Francis W. Davison, MD
1949
1950
1951
1952
1953
1954
1955
1956
1957
1958
1959
1960
1961
1962
1963
1964
1965
1966
1967
1968
1969
1970
1971
1972
1973
1974
1975
1976
1977
1978
1979
VICE-PRESIDENTS
Sam E. Roberts, MD
Murdock Equen, MD
Frederick T. Hill, MD
George J. Taquino, MD
Walter H. Hoover, MD
Daniel S. Cunning, MD
Howard W. D. McCart, MD
Walter P. Work, MD
Walter P. Work, MD
George S. McReynolds, MD
Daniel C. Baker, Jr, MD
Julius W. McCall, MD
Herbert W. Schmidt, MD
Herbert H. Harris, MD
James A. Harrill, MD
Paul G. Bunker, MD
Joel J. Pressman, MD
William A. Lell, MD
Arthur M. Olsen, MD
Oliver W. Suehs, MD
C. E. Munoz-MacCormick, MD
G. Edward Tremble, MD
John S. Knight, MD
Richard A. Rasmussen, MD*
Seymour R. Cohen, MD
Donald F. Proctor, MD
John E. Rayl, MD
Joyce A. Schild, MD
John P. Frazer, MD
James D. Baxter, MD
William R. Hudson, MD
1980
1981
1982
1983
1984
1985
1986
1987
1988
1989
1990
1991
1992
1993
1994
1995
1996
1997
1998
1999
2000
2001
2002
2003
2004
2005
2006
2007
2008
2009
2010
Bernard R. Marsh, MD
John E. Rayl, MD
Willard A. Fry, MD
Robert W. Cantrell, MD
Francis I. Catlin, MD
Henry J. Heimlich, MD
Robin T. Cotton, MD
LaVonne Bergstrom, MD
Donald B. Hawkins, MD
Thomas C. Calcaterra, MD
Mary D. Lekas, MD
Eiji Yanagisawa, MD
Eiji Yanagisawa, MD
Ellen M. Friedman, MD
Ellen M. Friedman, MD
W. Frederick McGuirt, MD
W. Frederick McGuirt, MD
Ellen M. Friedman, MD
Ellen M. Friedman, MD
Robin T. Cotton, MD
Jonathan E. Aviv, MD
Jonathan E. Aviv, MD
Jonathan E. Aviv, MD
Jonathan E. Aviv, MD
Jamie A. Koufman, MD
Jamie A. Koufman, MD
Jamie A. Koufman, MD
Ellen S. Deutsch, MD
Ellen S. Deutsch, MD
Ellen S. Deutsch, MD
Ellen S. Deutsch, MD
157
––––
*Served as President.
158
Officers
PRESIDENTS-ELECT
1969
1970
1971
1971
1972
1973
1974
1975
1976
1977
1978
1979
1980
Richard W. Hanckel, MD
John R. Ausband, MD
Robert E. Priest, MD
(April to Sept. ’71)
John S. Knight, MD
(Sept. ’71)
Gabriel F. Tucker, Jr, MD
Howard A. Anderson, MD
Walter H. Maloney, MD
Seymour R. Cohen, MD
Paul H. Ward, MD
James B. Snow, Jr, MD
Joyce A. Schild, MD
Loring A. Pratt, MD
M. Stuart Strong, MD
1981
1982
1983
1984
1985
1986
1987
1988
1989
1990
1991
1992
1993
1994
1995
Bernard R. Marsh, MD
John A. Tucker, MD
Frank N. Ritter, MD
William R. Hudson, MD
David R. Sanderson, MD
C. Thomas Yarington, Jr, MD
Robert W. Cantrell, MD
H. Bryan Neel III, MD, PhD
Gerald B. Healy, MD
Charles W. Cummings, MD
Lauren D. Holinger, MD
Haskins K. Kashima, MD
Eiji Yanagisawa, MD
Robert H. Ossoff, DMD, MD
Stanley M. Shapshay, MD
1996
1997
1998
1999
2000
2001
2002
2003
2004
2005
2006
2007
2008
2009
2010
Rodney P. Lusk, MD
W. Frederick McGuirt, MD
Paul A. Levine, MD
Ellen M. Friedman, MD
Robin T. Cotton, MD
Peak Woo, MD
Charles N. Ford, MD
Steven M. Zeitels, MD
Jonathan E. Aviv, MD
Gady Har-El, MD
Clarence T. Sasaki, MD
Jamie A. Koufman, MD
Andrew Blitzer, MD, DDS
Michael A. Rothschild, MD
Gregory Postma, MD
SECRETARIES
1928-1930
1931
1932
1933
1934-1939
1939-1948
1948-1953
Louis H. Clerf, MD
Richmond McKinney, MD
Edwin McGinnis, MD
Henry Hall Forbes, MD
Lyman Richards, MD
Paul H. Holinger, MD
Edwin N. Broyles, MD
1953-1960
1960-1965
1965-1970
1970-1974
1974-1978
1978-1984
1984-1990
F. Johnson Putney, MD
Daniel C. Baker, Jr, MD
John R. Ausband, MD
Walter H. Maloney, MD
Loring W. Pratt, MD
David R. Sanderson, MD
Lauren D. Holinger, MD
1990-1995
1995-1999
2000-2001
2002-2005
2005-2008
2009-
Rodney P. Lusk, MD
James A. Duncavage, MD
Charles N. Ford, MD
R. Kim Davis, MD
Peter J. Koltai, MD
Gregory A. Grillone, MD
1994-1999
2000-2002
2003-2005
2006-2009
2010-
Paul A. Levine, MD
Steven M. Zeitels, MD
Clarence T. Sasaki, MD
Gregory Postma, MD
Dana Thompson, MD
1989-1995
1995-2000
2000-2004
2004-2008
2008-
Stanley M. Shapshay, MD
Peak Woo, MD
Gady Har-El, MD
Michael A. Rothschild, MD
J. Scott McMurray, MD
TREASURERS
1928-1930
1930-1933
1933-1946
1946-1954
1954-1959
1959-1964
Henry B. Orton, MD
Waitman F. Zinn, MD
Robert M. Lukens, MD
Clyde A. Heatly, MD
Verling K. Hart, MD
Charles M. Norris, MD
1964-1969
1969-1977
1977-1982
1982-1985
1985-1990
1990-1994
Richard W. Hanckel, MD
John P. Frazer, MD
John A. Tucker, MD
C. Thomas Yarington, Jr, MD
Mary D. Lekas, MD
Robert H. Ossoff, DMD, MD
EDITORS
1928-1939
1939-1947
1947-1952
1952-1958
1958-1963
Ellen J. Patterson, MD
Louis H. Clerf, MD
Fred W. Dixon, MD
Francis W. Davison, MD
Stanton A. Friedberg, MD
1963-1968
1968-1972
1972-1977
1977-1983
1983-1989
Charles F. Ferguson, MD
Gabriel F. Tucker, Jr, MD
James B. Snow, Jr, MD
Frank N. Ritter, MD
Gerald B. Healy, MD