A First-in-Man Phase 1 Clinical Trial
Transcription
A First-in-Man Phase 1 Clinical Trial
Quality & Compliance A First-in-Man Phase 1 Clinical Trial A Tragic Ending leads to a New Guideline By Lorna Speid, PhD, RAC T Research Subject Safety Series Part 1 he injury of six healthy volunteer research subjects during a first-in-man phase 1 clinical trial at Northwick Park research center in 2006 sent shock waves throughout the pharmaceutical community and the general public worldwide. Many people, including those involved in the drug development process and clinical research, believed something was clearly amiss with the system for assessing clinical trials if this type of event could occur. Answers were demanded about what went wrong and what would need to change in the way clinical trials are conducted. In the aftermath of this event, there was an extensive investigation and report,1 and a new guideline2 was issued. Scientists have discussed at length the physiological and scientific issues involved.1,3,5 Project teams working on New Chemical Entities can learn important lessons from reviews and investigations of this event.1 This series of articles examines some of the issues regulatory affairs professionals on a project team must be able to handle on a day-to-day basis. It looks at the TeGenero clinical trial and its implications. This clinical trial resulted in significant harm to six healthy volunteers and may have changed the landscape of, at least, firstin-man clinical trial conduct for the foreseeable future. Part 1 of this three-part series reviews the guideline issued by EMEA as a result of this experience.2 Although the guideline was issued by European authorities, the principles are applicable anywhere first-in-man studies are conducted. The second article will examine lessons learned from this tragedy. In Part 3, the informed consent process will be discussed; we will consider how it must be designed and used to ensure research subjects are able to make truly informed decisions when considering participating in a clinical trial. Phase 1 Clinical Trials: TGN1412 (TeGenero) As is typical for clinical studies, several organizations were involved in conducting this clinical trial to evaluate TGN1412, a first-in-man monoclonal antibody. The sponsor—in this case 36 April 2008 TeGenero (Germany)—is ultimately responsible for the oversight of all aspects of the clinical trial. The drug product manufacturer was Boehringer Ingelheim, and the phase 1 unit based at London’s Northwick Park Hospital was run by PAREXEL, a Clinical Research Organization. TGN1412 was being developed for the treatment of rheumatoid arthritis, leukaemia and multiple sclerosis. It was administered by injection over a period of minutes, rather than a slow infusion over several hours, as may have been advisable. TeGenero filed a phase 1 clinical trial application for the compound with the UK Medicines and Healthcare products Regulatory Agency (MHRA) in early 2006. MHRA cleared the application without identifying any major issues. On 13 March 2006, eight healthy male volunteers participated in the double-blind, randomized, placebo-controlled phase 1 safety study of TGN1412. Two volunteers received placebo and the other six subjects received TGN1412. The study drug was a recombinantly expressed, humanized super-agonist monoclonal antibody.1,3,4,5 The six subjects receiving the active molecule were dosed within 10 minutes of one another. Within 90 minutes, all six experienced systemic inflammatory responses that caused their main body organs to collapse. This syndrome was characterized by a rapid induction of proinflammatory cytokines, accompanied by headache, myalgias, nausea, diarrhea, erythema, vasodilatation and hypotension. Within 12 to 16 hours following infusion, the subjects became critically ill with pulmonary infiltrates and lung injury, renal failure and disseminated intravascular coagulation. Severe and unexpected depletion of lymphocytes and monocytes occurred within 24 hours of infusion. Cardiopulmonary support (including dialysis), high-dose methylprednisolone and an anti–interleukin-2 receptor antagonist antibody were required. All six volunteers in the treatment group had multiorgan failure with an unknown mechanism and an unpredictable severity. They were admitted to the on-site critical care unit at Northwick Park and St. Mark’s Hospital, a National Health Service (NHS) hospital. Fortunately, all six research subjects survived, but it is anticipated that they will suffer longterm sequellae as a result of irreversible damage to their immune systems. They may have an increased lifetime susceptibility to various types of cancers. This is a tragedy for volunteers who were clearly healthy when they entered the clinical trial. The regulatory affairs professional plays an important role in ensuring that the EMEA guideline2 and Expert Report1 recommendations are implemented in first-in-man studies involving high-risk molecules. In fact, many of these recommendations would serve companies well in clinical programs in general. The new guideline2 is reviewed below; it reflects many of the findings from the investigation1 conducted following the TeGenero incident. Guideline Review The guideline encompasses many areas of development covered in more detail in other guidelines. For instance, it references International Conference on Harmonisation of Technical Requirements for Registration of Pharmaceuticals for Human Use (ICH) guidelines that must be considered during the development program.6-15 The guideline emphasizes that it is not applicable to gene and cell therapies. It was issued to address issues to be considered in working with novel compounds classified as high risk. After extensive investigation, the Committee on Human Medicinal Products (CHMP) determined that TeGenero had complied with all applicable regulatory guidelines. Since the incident occurred despite that compliance, additional guidance on high-risk molecules was deemed to be appropriate. The guideline captures important concepts and rules of drug development that many companies should be following routinely, regardless of the type of molecule being developed. Although the guideline does not cover much new material, it summarizes the types of issues to be considered in moving a potentially high-risk medicinal product through the development pathway and into the clinic. It recognizes the challenges of integrating the different decisions that need to be made about a compound moving through the early stages of development. The regulatory professional on the project team is often more familiar than most with these concepts, and should be confident enough to voice concerns when decisions do not take them into account. In this respect, the guideline will assist the regulatory professional make the arguments on these matters. In effect, it provides a crash course in good drug development practice, which will be particularly helpful to small companies or those new to drug development. For large pharmaceutical manufacturers, this guideline will be helpful, but that environment is likely to have more checks and balances, and therefore more opportunities to address the issues identified in the report and the guideline. Definition of High-Risk Molecules High-risk molecules are those that fulfill any of the following criteria: • present the potential for serious adverse reactions in first-in-man studies • target the immune system • have a mechanism of action that bypasses normal physiological control mechanisms, e.g., CD3 or CD28 supra agonists • are novel fusion proteins • are bi-specific antibodies • present difficulties in quantifying or predicting risks from animal studies and animal models due to speciesspecific responses, or difficulties in reproducing the human situation in animal studies Mode of Action of High-Risk Molecules There is concern about the mode of action and the difficulties of predicting the dose response curve for high-risk molecules. In situations where the molecule is able to act on many targets in an unpredictable way, constructing the dose finding curve may be difficult. This results in higher risks to research subjects for first-in-man studies. “Mode of action Consideration should be given to the novelty, plausibility and extent of knowledge of the proposed mode of action. This includes the nature and intensity (extent, amplification, duration, reversibility) of the effect of the active substance on the target and the type of dose response (linear, nonlinear, U-shaped, bell-shaped). Previous exposure of human beings to compounds that have related biological mechanisms should also be considered. For example, the following mechanisms could be considered as high risk: − A pleiotropic mechanism, e.g., leading to various physiological effects, or targets that are Regulatory Focus 37 ubiquitously expressed, as often seen in the immune system, − A mechanism that bypasses physiological control mechanisms, e.g., CD3 or CD28 (supra-) agonists.” [Extracted from Guideline 2] Predictive Value of Animal Studies The relevance of animal models is a particular concern for high-risk molecules, as is scale-up to appropriate species for Good Laboratory Practice toxicology studies. If the molecule does not produce the same dose effects in the species as it will in man, observed animal responses may not accurately predict the types of adverse responses that could be observed in man or the doses at which they might occur. These molecules must be considered high risk because it is impossible to predict the responses they will elicit in research subjects. More caution must therefore be exercised when conducting these studies. “Relevance of animal models The Sponsor should compare the available animal species to humans taking into account the target, its structural homology, distribution, signal transduction pathways and the nature of pharmacological effects. If available animal models are of limited relevance to study properly the pharmacological and toxicological effects of the medicinal product, it should be considered as highrisk.” [Extracted from Guideline 2] Demonstrating a Link Between Material in Toxicology Studies and Clinical Trials Demonstrating a link between the material used in the GLP toxicology studies and that used in clinical studies is critical. For instance, 38 April 2008 differences in the amounts and/or types of impurities in the material used in toxicology studies and the material used in clinical trials reduce the predictive ability of the GLP toxicology studies. One way of ensuring a link by manufacturing sufficient amounts of material for both the GLP toxicology studies and the first clinical study in the same batch. Selecting Doses for First-in-Man Studies Methods needed to determine first-in-man studies for high-risk molecules may be different from those used to determine the starting dose for other molecules. The guideline2 and the official investigation report1 suggest some methods for calculating starting doses. “4.3.6 Calculation of the first dose in man In general, the calculation of the first dose in man is based on No Observed Adverse Effect Level (NOAEL) determined in nonclinical safety studies performed in the most sensitive and relevant animal species, adjusted with allometric factors or on the basis of pharmacokinetics. The relevant dose is then reduced/adjusted by appropriate safety factors according to the particular aspects of the molecule and the design of the clinical trials. For high-risk medicinal products, an additional approach to dose calculation should be taken. The use of ‘Minimal Anticipated Biological Effect Level’ (MABEL) approach is recommended. The MABEL is the anticipated dose level leading to a minimal biological effect level in humans. Safety factors are usually applied for the calculation of the first dose in man from MABEL. The calculation of MABEL should utilise all relevant in vitro and in vivo available information from pharmacodynamic/ pharmacokinetic data such as: i) receptor binding and receptor occupancy studies in vitro in target cells from human and the relevant animal(s) species and in vivo in the relevant animal species. ii) concentration-response curves in vitro in target cells from human and the relevant animal(s) species and dose response in vivo in the relevant animal species. iii) exposures at pharmacological doses in the relevant species. The above data should be integrated in a PK/PD modelling approach for the determination of the MABEL. In order to further limit the potential for adverse reactions in humans, safety factors should be applied in the calculation of the first dose in man from the MABEL. These should take into account criteria of risks such as the novelty of the active substance, its biological potency and its mode of action, the degree of species specificity, and the shape of the dose-response curve. The safety factors used should be justified. When the methods of calculation (e.g. NOAEL, MABEL) give different estimations of the first dose in man, the lowest value should be used.” [Extracted from Guideline 2] Level of Acceptable Risk Research subjects must not be exposed to undue risk. Preclinical work must demonstrate an acceptable level of risk. This requirement is not new; all clinical research carries some risk. However, healthy volunteers in a phase 1 trial will not experience any benefit from taking the drug, therefore the level of risk must be very small. Regulatory Standards and Compliance Good Manufacturing Practices (GMPs) must be followed in manufacturing the material to be administered to research subjects. For US-based companies following a graded GMP strategy as development progresses, discussions with European regulatory authorities may be required to ensure that GMP standards being followed for the phase 1 studies are acceptable. The guideline emphasizes the need to achieve a high level of quality characterization before the phase 1 study. This implies that analytical methods will be developed and validated prior to the start of toxicology studies. The molecule or compound will also require significant scientific characterization in advance of the first-in-man phase 1 study. Companies focused on developing biological agents may tend to try following a graded approach in the application of GMPs and Good Laboratory Practices. Had TeGenero not been highly compliant in these areas, it would have been difficult to determine whether the events that occurred were due to the quality of the material used, immune responses or other factors. The apparent high level of compliance with GLP, GMP and GCP standards made it easier to identify the pharmacological issues that may have contributed to the adverse events. Target Organ Toxicity Elucidation Good Laboratory Practice (GLP) toxicology studies should demonstrate target organ toxicities to enable the patient study(-ies) to include appropriate safety monitoring. This may be difficult if the compound cannot be administered to animals in large amounts, e.g., if humanized molecules are used. In such a case, discussion with the regulatory agency may be appropriate to determine whether special animal toxicology models should be developed to study mechanisms of toxicity before first-in-man studies can begin. Choice of Research Subject Population The population to be studied should be selected carefully. For a novel immunomodulator, patients may be more appropriate than healthy volunteers. A healthy immune system may overreact to the administration of immunomodulators, resulting in collapse or attack on the body systems. Facilities for Conducting Studies of High-Risk Molecules The guideline2 and the investigation1 highlighted the fact that the TeGenero study was conducted in a hospital facility with expert emergency physicians and other staff who may have saved the participants’ lives. First-in-man studies should take place in appropriately staffed and equipped medical facilities, even for healthy volunteer studies. Healthy volunteer units without medical facilities in close proximity to the research unit should be avoided for studies involving high-risk medicinal products. Dosing High-Risk Molecules It is important to proceed cautiously with dose escalation after starting at the lowest feasible dose. Adequate intervals must be left between dosing research subjects. In the TeGenero study all subjects were dosed within 10 minutes of one another. This left little room for dose adjustment or study termination when problems became evident approximately 90 minutes after dosing began. High-risk molecules need to include rules for stopping dosing (stopping rules) that should be pre-agreed with the health authority and activated as conditions require. Stopping rules should detail appropriate steps to be taken in the event of various eventualities. Regulatory Focus 39 Safety Monitoring During the Study Research subjects in whom high-risk molecules are being studied should remain on site for the study’s duration. They also should be monitored after leaving the facility for a sufficient period of time after dosing to detect any delayed responses. It may be appropriate to establish an independent drug safety monitoring board to oversee the safety of studies involving highrisk medicinal products. A monitoring plan should be developed for the study that includes a risk management plan, which should identify the likely risks and how each of them will be mitigated during the clinical trial. This monitoring plan should be communicated to study physicians and nurses, and to emergency personnel who will be on standby during the clinical trial. Mechanisms for safety reporting should be in place before the study begins. Conclusion As the pharmaceutical industry and research institutions seek to address as yet unmet medical needs, molecules of a type not previously studied in man may be created. These molecules may need to be 40 April 2008 classified as high risk, depending upon the ease with which they can be studied in animals, as well as the type of reactions they may elicit in the human body. Where they are classified as high risk, care must be taken to ensure that risks to research subjects are appropriately controlled. Adherence to the current guideline will help reduce the potential for another incident like TeGenero. It is hoped the guideline will be dynamic since additional guidance may be needed or updates made as more experience is gained with high-risk molecules. Although the guideline was issued in Europe, and the US and Japan do not yet have their own versions, the recommendations it contains may be relevant for studies conducted elsewhere. On the contrary, because the pharmaceutical industry is involved in global product development, what happens in Europe impacts drug development programs designed in the US and elsewhere. REFERENCES 1. Expert Scientific Group on Phase One Clinical Trials. Final Report, 30 November 2006. 2. Guideline for Requirements for First in Man Clinical Trials for Potential High Risk Medicinal Products. Committee for Medicinal Products for Human Use (CHMP).EMEA /CHMP/SWP/28367/2007. Corr.23 May 2007. Suntharalingam G, Perry MR, Ward S, Brett SJ, Costello-Cortes A, Brunner MD, Panoskaltsis N. “Cytokine Storm in a Phase 1 Trial of the Anti-CD28 Monoclonal Antibody TGN1412.” N Engl J Med, 2006; 356(10): 1018-1028. 4. TGN1412 investigator’s brochure. Wurzburg, Germany: TeGenero Immunotherapeutics, 2005. Accessed 11 August 2006. www.mhra.gov.uk/home/ idcplg?IdcService=GET_FILE&dDocName=CON202351 8&RevisionSelectionMethod=LatestReleased. 5. Investigations into adverse incidents during clinical trials of TGN1412. London: Medicines and Healthcare products Regulatory Agency (MHRA), 2006. Accessed 11 August 2006. www.mhra.gov.uk/home/ idcplg?IdcService=GET_FILE&dDocName=CON202382 1&RevisionSelectionMethod=LatestReleased . 6. “Non-Clinical Safety Studies For The Conduct Of Human Clinical Trials For Pharmaceuticals.” (ICH M3), CPMP/ICH/286/95. 7. “Preclinical safety evaluation of biotechnology-derived pharmaceuticals.” (ICH S6) CPMP/ICH/302/95, 8. “The Non-clinical Evaluation of the Potential for delayed Ventricular Repolarization (QT Interval Prolongation) by Human Pharmaceuticals.” (ICH S7B) CPMP/ICH/423/02. 9. “Safety pharmacology studies for human pharmaceuticals.” (ICH S7A) CPMP/ICH/539/00. Quality aspects. 10. EUDRALEX—Volume 4—Medicinal Products for 3. 11. 12. 13. 14. 15. Human and Veterinary Use: Good Manufacturing Practice. “Annex 13: Manufacture of Investigational Medicinal Products.” Guideline on Virus Safety Evaluation of Biotechnological Investigational Medicinal Products—Draft. EMEA/ CHMP/BWP/398498/2005-corr. Guideline on the Requirements to the Chemical and Pharmaceutical Quality Documentation concerning Investigational Medicinal Products in Clinical Trials. CHMP/QWP/185401/2004. Guideline for Good Clinical Practice. (ICH E6), CPMP/ICH/135/95. General Considerations for Clinical Trials, (ICH E8) CPMP/ICH/291/95. EUDRALEX—Vol 10—Clinical trials. In particular: Chapter I: Application and Application Form and Chapter II: Monitoring and Pharmacovigilance. AUTHOR Lorna Speid, PhD, RAC, is President of Speid & Associates Inc. (www.sndtm.com) a regulatory affairs and drug development consultancy based in San Diego. She can be reached at [email protected] or at +1 858 793 1295. Regulatory Focus 41