PERSPECTIVES
Transcription
PERSPECTIVES
PERSPEC TIVES 10. British Broadcasting Corporation. Moorfields hospital carries out world’s first gene therapy operation to cure blindness. <http://www.bbc. co.uk/pressoffice/pressreleases/stories/2007/05_ may/01/moorfields.shtml> (1 May 2007). Accessed 1 July 2007. 11. Piquette-Miller, M. & Grant, D.M. The art and science of personalized medicine. Clin. Pharmacol. Ther. 81, 311–315 (2007). 12. Guttmacher, A.E. & Collins, F.S. Realizing the promise of genomics in biomedical research. JAMA 294, 1399–1402 (2005). 13. National Cancer Institute and National Human Genome Research Institute. The Cancer Genome Atlas <http://cancergenome.nih.gov/index.asp>. Accessed 1 July 2007. 14. Food and Drug Administration. Guidance for Industry: Pharmacogenomic Data Submissions <http://www.FDA.gov/cber/gdlns/pharmdtasub. htm> (March 2005). Accessed 1 July 2007. Beyond Genomics CT Dollery1 The sequencing of the human genome has already had an enormous impact on medicine, particularly with single-gene changes that predispose to a serious disease such as cystic fibrosis or the overexpression of Her2 in about one-third of breast cancers. Genetic technology has led to some very important therapeutic innovations, including the use of imatinib mesylate (Gleevec) in BCR-ABL chronic myeloid leukemia and of trastuzumab (Herceptin) in Her2-positive breast cancer, but the much anticipated explosion of new effective treatments has been more modest than expected. Scientific meetings are now held regularly to bemoan the decline in registrations of new drugs with regulatory agencies such as the Food and Drug Administration and to explore the reasons. Many explanations have been put forward, including excessively stringent regulation, limitations of highly automated industrial drug discovery methods, and unimaginative clinical trials. All these potential explanations have their advocates, but the realization is slowly dawning that the biggest problem is lack of good targets because of the limited understanding of the etiologic mechanisms of most common diseases. Silver bullet or carronade? Cancer is the ultimate genetic disease, and sequencing the human genome would have been justified purely for the insights it gave into oncology. But apart from virology and cancer, genetics has not thus far been nearly as helpful as was expected in other disease areas. A few have drawn the uncomfortable conclusion that the underlying explanation doi:10.1038/sj.clpt.2007.6100363 366 Beyond genomics? for the lack of success in defining new targets may be that most common diseases are driven by the coincidence of multiple factors without one being dominant. A quote from a recent article in Nature describing a large genetic study of seven common diseases serves to illustrate this point: “For any given trait there will be few (if any) large effects, a handful of modest effects, and a substantial number of genes generating small or very small increases in disease risk.”1 If that is the case, the existing paradigm of drug discovery and development requires radical rethinking, as does the concept of personalized medicine based solely on genetics. Exit the silver bullet and enter the carronade (the carronade was a short-barrel, large-diameter cannon that could be loaded with a canister of 500 musket balls). Nowhere is the lack of progress in understanding mechanisms underlying the etiology and maintenance of most common disease processes felt more keenly than in the pharmaceutical indus- 1GlaxoSmithKline, Harlow, UK. Correspondence: CT Dollery ([email protected]) try. The cascade of new “druggable targets” disclosed by the sequencing of the human genome (>300 G protein–coupled receptors, >500 kinases, and a similar number of proteases)2 has had a much more limited impact than early enthusiasts anticipated. The attrition rate of pharmaceutical projects aimed at these novel targets is very high (estimated to be ∼95% failure rate for drugs aimed at novel targets), and one of the most common reasons given is “lack of efficacy.” In many cases this is a misnomer: the drug had the anticipated pharmacological action, but the effect on the disease mechanism was small or nonexistent. A better way of describing the result would be “poor choice of target.” Many human diseases reflect a disorder in physiologic processes such as blood pressure, body weight, or inflammation that are known to involve the interaction of many complex control loops and to respond to some degree to a variety of pharmacologic agents and environmental factors. It is highly unlikely that the genome of the average greater than third-generation American has undergone much change in the last 75 years, and the same proportion must carry the FTO gene,3 but the incidence of obesity and type II diabetes has increased markedly. Agents as diverse as diuretics, α- and β-adrenergic antagonists, angiotensin-converting enzyme inhibitors, angiotensin II antagonists, and calcium L channel blockers lower blood pressure in hypertension to some degree, but none of them cures it, and disorders of these control systems have not been identified as playing a major function in its etiology. Against this background it should have been anticipated that the genetic influences in these conditions were likely to be complex and multifactorial. The optimists argue that all that will be needed is to subdivide the existing broad phenotype into several new diseases in each of which only one or two gene polymorphisms will be major etiologic factors. The pessimists (realists?) argue that everyone will have a mix of environmental and genetic risk factors, and although the mix will vary, multiple processes will be at work in almost all. VOLUME 82 NUMBER 4 | OCTOBER 2007 | www.nature.com/cpt PERSPEC TIVES Renaissance of integrative physiology and pharmacology Ultimately the choice of drug targets must rest on their ability to modify a disease process, but a critical step toward that objective is to understand the normal physiologic functions of target molecules. A large part of the current problem in drug discovery is that the productivity of genomics technology, for a time, far outran biological research capacity to translate this information into insights about the role of a gene product in normal physiology. This imbalance is beginning to be addressed by using genomic techniques such as quantitative gene expression (mRNA), gene knockout, tissue-specific gene inactivation (CreLox), small interfering RNA (siRNA) gene silencing, and monoclonal antibodies combined with accurate physiologic measurements to understand the tissue distribution and physiologic function of gene products. In humans it has been necessary to rely more on experiments of nature in the shape of genetic polymorphisms. One consequence of the renaissance in experimental physiology has been to reveal a severe shortage of physiologists trained in the study of integrated systems in intact animals and humans. The importance of these developments to pharmacology should not be underestimated. Pharmacology grew out of physiology, and a renaissance of integrative physiology should be matched by a similar commitment to integrative pharmacology both preclinically and clinically. A word of caution is in order, though, and it pertains to an additional reason for quality clinical pharmacology. The function in humans of a protein that has a close (or identical) animal homologue may differ substantially, as was shown with the CD28 antibody TGN1412 that was relatively benign in cynomolgus monkeys but caused a devastating cytokine storm in humans.4 Disease models in preclinical species as predictors for human pathophysiology are another story. They worked well for deficiency diseases, bacterial infections, hypertension, and some types of inflammation. In an era in which the major disease targets are conditions such as virus infections, cancer, dementia, schizophrenia, and osteoarthritis, their value is much more limited. Whether these models can be made more useful by knocking in human genes to preclinical species remains to be seen, in that reproducing the complexity of human disease etiology in a mouse will be of daunting complexity. For the present the chief experimental animal for understanding human pathophysiology is the human, and clinical pharmacology has an important role to play by using drugs to illuminate mechanisms. Aghast at the complexity? Sidney Brenner, a Nobel laureate, once said that the problem of biology is not to stand aghast at the complexity but to conquer it.5 It is also a fitting challenge for clinical pharmacologists faced with the challenge of making drug therapy safer and more effective in the real world. Where should they start? One obvious answer is that they cannot do it alone. A multidisciplinary team approach will be essential, ranging from biostatisticians to practitioners of family and specialist medicine, but the two key components are knowledge of the drug action in humans and detailed knowledge of the patient taking the drug. The former is, or should be, the purview of the clinical pharmacologist. The latter requires a strong background in clinical medicine either through personal knowledge and training as part of a clinical pharmacology program or with a committed clinical collaborator. Either way, a deep understanding of the often complex clinical situation is necessary. If clinical pharmacologists are to fulfill a front-line role in the study of drugs in patients and make a major contribution to personalized medicine, the retreat into the laboratory and to studies largely confined to healthy volunteers—not patients—will have to be reversed. Back to the clinic Some clinical pharmacologists will participate directly in clinical care, because they also have trained in a specialty such as oncology, psychiatry, or cardiology. For them, direct access to patients is an obvious advantage. Others will participate in clinical research in a more collaborative role, but even if they are not themselves medically trained, the acquisition of a thorough knowledge of the disease and patients under study will greatly strengthen their CLINICAL PHARMACOLOGY & THERAPEUTICS | VOLUME 82 NUMBER 4 | OCTOBER 2007 contribution to clinical research and the willingness of clinical colleagues to collaborate. Where should they start? Investigation of outliers Pharmaceutical companies sponsoring research on their compounds, regulatory authorities considering whether to license them, and purchasing agencies considering whether to authorize their use in local formularies pay most attention to the magnitude of benefit and its statistical significance for the whole group of patients in a clinical trial. For the clinical investigator looking for clues, a better place to start is to investigate outliers. These could be patients suffering a serious adverse reaction to a medicine that is safe in the great majority. Several important but rare polymorphisms of drug metabolism have been discovered in this way, including the dihydropyrimidine dehydrogenase polymorphism and 5-fluorouracil (5-FU) toxicity.6 But this is not the only mechanism of severe 5-FU toxicity. The interaction between the anti–herpes zoster drug sorivudine (5-bromovinyl-ara uracil) and standard doses of 5-FU, which resulted in 18 deaths, was another salutary example that might have been foreseen.7 The outlier approach need not be confined to adverse reactions; it can also be applied to clinical trial data to investigate, say, the top 5% to 10% of excellent responders and the bottom 5% to 10% who had little or no response. Lessons learned from outliers often prove to have applications to the mass of patients who are not outliers. Design of clinical trials The whole fabric of evidence-based medicine and the Cochrane database rests on large, well-designed, randomized controlled clinical trials. Historically, clinical pharmacologists have only a limited impact on the design of such trials. This may account for the relative neglect of important issues such as dose-response or concentration-response, for both efficacy and side effects, in many large trials. Fortunately, the situation is changing with the growing use of pharmacokinetic-pharmacodynamic (PK/PD) analyses in assessing individual differences in response. This is not merely a matter of combining pharmacodynamic and pharmacokinetic data, 367 PERSPEC TIVES because the relationship is often far from a simple log-dose response curve and may be quite different for efficacy parameters and adverse effects more directly related to the pharmacology. A knowledge of the clinical pharmacology and its relationship to the disease mechanism is essential both in the design and the analysis of clinical trials to secure the best yield of useful information and the most valuable interpretation of the data. Clinical pharmacologists are, or should be, much more interested in accurate recording of symptomatic side effects as part of the spectrum of pharmacologic activity than the epidemiologists, statisticians, and clinician specialists who usually have their main attention devoted to demonstrating efficacy. One important additional skill the clinical pharmacologist can bring is to increase the accuracy of individual measurements in clinical trials. Most measurements made in large clinical trials are relatively imprecise. The objective is to show a significant effect for the group as a whole. To gain useful information for personalized genetic studies, individual PK/PD analysis, and the like, the accuracy of individual measurements will have to be improved. It is already an issue in adaptive trial designs. It will become even more critical as we move more toward factorial designs of multidrug combinations in relatively small groups of patients. Personalized medicine When a subway train stops in London, overseas passengers are often amused when the station loudspeakers start playing a message to “mind the gap” between the train and the curved platform. Two recent edited conversations illustrate the gap in understanding of the background to personalized medicine. The first was with a senior, very experienced internist who had just finished reading a journal article about personalized medicine. She mused aloud that she thought she had practiced personalized medicine throughout her professional life. She took a careful medical, family, and social history including a detailed account of any current treatment or recent past therapy, alcohol and tobacco intake, and so on. Most of her patients were over 60 years old, and it was a rarity that they were being 368 treated for only one medical condition, so most were on several drugs. She dreaded the moment when an elderly patient produced a bag containing a large assortment of prescription and nonprescription drugs without a clear recollection of which he or she was still taking and when. Her greatest contribution to personalized medicine was often to have a patient stop taking at least half of this large assortment of medicines. The second conversation was with a younger nonclinical scientist at a pharmacokinetics meeting. He tackled me, apparently as an elderly symbol of medical conservatism, to know why all the discoveries about genetics polymorphisms of the drug-metabolizing enzymes had had so little influence on medicine. Why didn’t physicians routinely assess the CYP2D6 polymorphism before prescribing metoprolol, paroxetine, or codeine and measure at least CYP2C9 polymorphism, let alone VKORC1, before choosing a starting dose of warfarin? Perhaps, he implied, the coming of the Amplichip CYP450 (ref. 8) gene array chips for drug metabolism would drag the medical profession out of the therapeutic dark ages. Although the gap between the London subway train and the platform is rarely more than a few inches, the gap these two conversations illustrate is much wider, and we must find ways to bridge it. Both parties have some right on their side, but the reductionist approach has important limitations in very complex realworld situations. Testing hypotheses in personalized medicine: go for big effects It is relatively easy to propose strategies for improving personalized medicine but much harder to test and implement them. The physician whose treatment advice to a new patient starts with “I want you to stop smoking, halve your alcohol intake, and lose 30 pounds” soon learns from direct personal experience that this will not work, in a small number of patients. In most cases, it will be more difficult than this simple example to test new strategies in personalized medicine and will require large-scale testing in clinical trials before anyone will take the new ideas seriously. Those familiar with calculating the numbers required in clinical trials will know how large these can be if the intention is to demonstrate superiority for a new medicine that is expected to have, say, a 10% advantage over an existing medicine for a common disease. Similar considerations will apply to testing personalized medicine strategies. There must be good grounds, based on careful modeling, for anticipating a sizable effect. For example, a major metabolic route for metoprolol is via CYP2D6, and in patients with polymorphisms that reduce its function, plasma concentrations are higher, sometimes much higher. However, within groups with similar genotypes concentrations also vary widely. To convince a health-care purchaser to use CYP2D6 genotyping routinely when prescribing metoprolol would probably require the design of a trial to show not just that the concentration achieved was more consistent but that this was sufficient to produce a worthwhile clinical improvement in the control of angina or hypertension. Given the high variability within genotypes this would be difficult, although not necessarily impossible. But it makes the point that the first thing to do when considering a new strategy for personalized medicine is to estimate the contribution of the factors under study to the variance in response and concentrate on those that have the potential to make a big difference. Step one is to estimate the effect on the variance in the pharmacological mechanism but step two is to model the effect that change in pharmacology would have on the therapeutic response. Both are areas in which the clinical pharmacologist ought to be able to make a major contribution. Integrating knowledge, building models At the battle of Trafalgar a canister of musket balls from the 68-pound carronade on Horatio Nelson’s flagship killed or wounded hundreds of men operating the guns on the French flagship. Moments later, a French sharpshooter fatally wounded Nelson with a single musket ball. Once battle was underway, which was the more significant shot? Nelson died of his wound, but the French flagship was captured. For more than 50 years medical educators have taught their students to abhor blunderbuss therapy and choose the elegant, carefully selected, single-drug VOLUME 82 NUMBER 4 | OCTOBER 2007 | www.nature.com/cpt PERSPEC TIVES intervention. But we now live in an era when well-known epidemiologists advocate a “polypill” for preventing heart disease9 and patients with many common diseases (hypertension, diabetes, cancer) are routinely treated with more than one drug. The wide-ranging implications of these changes have received little attention from clinical pharmacologists apart from studies of interactions in drug metabolism. Yet logical, well-conceived drug combinations may prove to be the best solution to diseases driven by multiple etiologic factors in complex control systems. Patents are already appearing for devices that are intended to dispense individualized “polypills.” Some of the drivers for a new era of scientifically credible polypharmacy will be genetic and some environmental; often the end result will be determined by the play of one on the other. This may turn out to be the real personalized medicine. The classical reductionist approach of science to a complex problem is to stabilize as many variables as possible and alter one, judged to be important, to see what happens to another that has not been stabilized. In the early days of experimental physiology and pharmacology of the cardiovascular system, this often worked brilliantly. But the major unmet clinical needs of today (osteoarthritis, diabetes, lung cancer, chronic obstructive pulmonary disease, schizophrenia, dementia, etc.) present a more difficult challenge and have not thus far proved particularly amenable to traditional experimental approaches. Nor has genetics identified single factors making a major contribution to the variance. An alternative approach is to focus efforts on the pathways and the control systems that influence the disease state under study to identify nodes particularly suitable for intervention. It is a complex task but potentially very rewarding. Might some of those compounds directed at that G protein–coupled receptor or this kinase, and discarded as ineffective, prove to be useful as part of a carefully chosen combination strategy? Might a mutation in one part of a control system that is not readily accessible to intervention be circumvented by modulating another control loop with which it interacts? These data can be used as a basis for building a mathematical model of the dis- ease that will require successive iterations between model and experiment to test it. This approach, now termed systems biology, is bound to require a great deal of experimental work in humans, perhaps using tool compounds not fully optimized as drugs and very sophisticated measuring techniques to assess effects over relatively short periods of drug administration. Populations are our paymasters and our laboratory In an increasingly cost-conscious healthcare world, the benefit and risk of therapeutic interventions are being closely studied. Bodies such as the National Institute of Health and Clinical Excellence in the United Kingdom10 have set a notional maximum the British National Health Service will pay for one additional quality-adjusted year of life (currently said to be ∼$60,000). Some purchasers argue that they should pay only for medicines that have been demonstrated to benefit the individual patient. These are powerful forces and will mobilize pressure to devise better methods of identifying responders and nonresponders among the generality of patients, not just the “sanitized” individuals recruited into clinical trials. Clinical pharmacologists ought to be in the forefront of work on these areas alongside health economists, but in doing so they will have to face some new realities. In the United States it is estimated that more than half of patients do not comply with prescription instructions and almost 20% of prescriptions are never filled.11 The frequency of refilling prescriptions suggests that many patients who do take their medicine fairly regularly are taking appreciably less than the prescribed dose. Many of the patients will be elderly and under treatment for more than one disease, each with more than one drug. The noise level in the data will be very high, but the problem is not insurmountable. The dramatic impact that new drugs have had, particularly on cardiovascular disease and HIV, shows that big signals can penetrate the noise. Very large patient databases will become important tools. At present these are usually mined for safety information on a single drug, but there is considerable potential to study the effect of combinations, both planned and incidental, on efficacy. Statis- CLINICAL PHARMACOLOGY & THERAPEUTICS | VOLUME 82 NUMBER 4 | OCTOBER 2007 tical and epidemiologic expertise will be essential, but so will knowledge of clinical pharmacology and internal medicine. Novel hypotheses generated in this way will require modeling using systems biology approaches, and sophisticated testing in the clinic. A new dawn Gloom about the present situation of drug discovery has been much exaggerated. Its origin was an unrealistic assessment of the time scale and biological complexity involved in applying the new knowledge from the sequencing of the human genome. In retrospect, sequencing was the easy part. But we know more about biology today than ever before. Our ability to make rapid and accurate diagnoses of human disease has made enormous progress powered, particularly, by imaging technology. The fusion of genetic and molecular techniques with integrative physiology and pharmacology is beginning to pay off with exciting new discoveries. These will translate into much better understanding of normal control systems and the way pharmacologic agents interact with them, for example, the varied ways that different anesthetic agents acting on the γ-aminobutyric acid receptor can interfere with memory12 or the marathon mouse construct with peroxisome proliferative activated receptor δ. Our understanding of the mechanisms responsible for the etiology and progression of human disease (not necessarily the same) has lagged somewhat, but many of the tools needed to advance experimental medicine and clinical pharmacology now exist. We are not “beyond genomics” but over the hump of unrealistic optimism. There will be major payoffs, it may take longer than we hoped, and it will require an enormous amount of hard work and large sums of money, but we must not let the patients down who so badly need better medicines. CONFLICT OF INTEREST The author declared no conflict of interest. © 2007 ASCPT 1. 2. The Wellcome Trust Case Control Consortium. Genome-wide association study of 14,000 cases of seven common diseases and 3,000 shared controls. Nature 447, 661–678 (2007). Hopkins, A.L. & Groom, C.R. The druggable genome. Nat. Rev. Drug Discov. 1, 727–730 (2002). 369 PERSPEC TIVES 3. 4. 5. 6. 7. Frayling, T.M. et al. A common variant in the FTO gene is associated with body mass index and predisposes to childhood and adult obesity. Science 316, 889–894 (2007). Suntharalingam, G. et al. Cytokine storm in a phase 1 trial of the anti-CD28 monoclonal antibody TGN1412. N. Engl. J. Med. 355, 1018– 1028 (2006). Academy of Medical Sciences and The Royal Academy of Engineering. Systems Biology: A Vision for Engineering and Medicine <http:// www.raeng.org.uk/policy/engagement/pdf/ Systems_Biology_Report.pdf> (February 2007). Wei, X., McLeod, H.L., McMurrough, J., Gonzalez, F.J. & Fernandez-Salguero, P. Molecular basis of the human dihydropyrimidine dehydrogenase deficiency and 5-fluorouracil toxicity. J. Clin. Invest. 98, 610–615 (1996). Okuda, H. et al. Lethal drug interactions of sorivudine, a new antiviral drug, with oral 5fluorouracil prodrugs [corrected and republished from Drug Metab. Dispos. 25, 270–273 1997, PMID: 9029059]. Drug Metab. Dispos. 25, 270–273 (1997). 8. Jain, K.K. Applications of AmpliChip CYP450. Mol. Diagn. 9, 119–127 (2005). 9. Wald, N.J. & Law, M.R. A strategy to reduce cardiovascular disease by more than 80%. BMJ 326, 1419 (2003). 10. National Institute for Health and Clinical Excellence <http://www.nice.org.uk>. 11. Krueger, K.P., Felkey, B.G. & Berger, B.A. Improving adherence and persistence: a review and assessment of interventions and description of steps toward a national adherence initiative. J. Am. Pharm. Assoc. 43, 668–679 (2003). 12. Orser, B.A. Lifting the fog around anesthesia. Sci. Am. 296, 54–61 (2007). The mitochondrial PTP as a target for modulating cell death Targeting Cell Death DJ Hausenloy1 and L Scorrano2 Functional consequences of myocardial or cerebral infarction are the result of excessive cell death. It is patent that preventing cell death is the therapeutic goal in any ischemia-reperfusion setting. Mitochondria amplify apoptotic cascades and have emerged as crucial organelles in ischemia-reperfusion. Changes in mitochondrial inner membrane permeability and in the morphology of the organelle are regulated, perhaps interconnected processes that are starting to emerge as novel therapeutic targets for reducing cell death induced by ischemia-reperfusion. Apoptosis is a genetic program of cell death, conserved among all metazoans, required for normal embryonic development, and essential to maintain tissue homeostasis by offsetting cell division. Malignant cells are able to evade death and proliferate by counteracting endogenous death mechanisms. Therefore, promoting death of tumor cells is the underlying aim of most anticancer therapies. Conversely, cell death induced by pathological stressors such as ischemiareperfusion as well as environmental toxins and genetic lesions predisposing to neurodegeneration is clearly detrimental. Ischemia-reperfusion in the context of a myocardial or cerebral infarction has seri- ous clinical repercussions; the therapeutic aim is therefore to prevent or reduce cell death. This limits the extent of the infarct and improves clinical outcomes, related to the size of the infarcted area. In an acute myocardial infarction (AMI), the prolonged acute myocardial ischemia results in cardiomyocyte death. Because the duration of acute myocardial ischemia is a major determinant of infarct size, removal of the thrombotic occlusion and restoring coronary artery flow is the most effective strategy for salvaging viable myocardium. Myocardial reperfusion is currently achieved by thrombolytic therapy or primary percutaneous coronary intervention, 1The Hatter Cardiovascular Institute, University College London Hospital, London, UK; 2Dulbecco-Telethon Institute, Venetian Institute of Molecular Medicine, Padova, Italy. Correspondence: L Scorrano (lscorrano@dti. telethon.it) doi:10.1038/sj.clpt.2007.6100352 370 strategies that have markedly improved the clinical outcome following an AMI. However, the restoration of coronary blood flow paradoxically induces further cardiomyocyte death, thereby reducing the overall benefits of myocardial reperfusion—a phenomenon termed lethal reperfusion injury. Mounting pharmacological and genetic evidence indicates that a critical mediator of this form of cell death is the mitochondrial permeability transition pore (PTP), whose opening early in the course of myocardial reperfusion disrupts mitochondrial function and activates the postmitochondrial apoptotic cascade. The PTP is a nonselective, tightly regulated high-conductance channel of the inner mitochondrial membrane.1 Its opening collapses the mitochondrial membrane potential, depletes the mitochondrial NADH pool, and uncouples oxidative phosphorylation, leading to a vicious circle of ATP hydrolysis and depletion, release of caspase cofactors and activators, and ultimately to cell death.2 The actual composition of the PTP is a matter of debate. Previous models that included the voltage-dependent anion channel (VDAC) of the outer membrane and the adenine nucleotide translocase of the inner membrane have been challenged by genetic studies, suggesting that neither VDAC3 nor adenine nucleotide translocase4 is an obligatory component of the PTP. In contrast, the matrix protein cyclophilin-D (CypD) has been found to be a strong facilitator of PTP opening, and its ablation results in increased resistance to the PTP inducer Ca2+, yet not to the absence of PTP.5 Cells lacking CypD were found to be resistant to ischemiareperfusion damage, both in the brain and in the myocardium; 6,7 on the other hand, the response to classic intrinsic apoptotic inducers was unaffected, a result that was interpreted to exclude the participation of the PTP in the core mitochondrial death pathway of apoptosis.8 A detailed discussion on the precise role of PTP in the mitochondrial phase of apoptosis is beyond the scope of this article. Nevertheless, this genetic evidence strongly suggests that PTP is crucially involved in cell death during ischemia-reperfusion damage, VOLUME 82 NUMBER 4 | OCTOBER 2007 | www.nature.com/cpt