challenges in developing therapies for rare diseases including pachyonychia congenita

5
Challenges in Developing Therapies for Rare Diseases Including Pachyonychia Congenita Roger L. Kaspar Transderm and SomaGenics, Inc., Santa Cruz, California, USA The ability to attract sufficient resources to effectively develop therapeutics for rare diseases is a daunting task. This review summarizes existing resources for rare diseases and discusses some of the challenges and strategies associated with developing therapies for small patient populations with an emphasis on pachyonychia congenita. Key words: orphan disease/pachyonychia congenita/rare disease J Investig Dermatol Symp Proc 10:62 –66, 2005 A rare (orphan) disease is defined under the United States Orphan Drug Act amendment (Orphan Drug Act, P.L. 97–414 (1983); Health Promotion and Disease Prevention Amend- ments, P.L. 98–551 (1984)) as a disorder that generally af- fects less than 200,000 individuals ( 0.07% of the US population). The definition is extended to disorders that af- fect greater numbers of individuals for which drug devel- opment is not likely, due to the expectation that the sales of such a drug would not be sufficient to recover development costs. Similar rare disease definitions are used by other countries including the European Community, Japan, Singapore and Australia (see the websites, http://www. rare-cancer.org/rare-diseases.html and http://www.europarl. eu.int/stoa/publi/167780/chap5_en.htm). Approximately 6,000 rare diseases have been identified and a list is maintained by the Office of Rare Diseases (ORD) at the National Institutes of Health (NIH) (http://ord.aspensys. com/asp/diseases/diseases.asp). Some of the listed rare diseases are well-known, such as sickle cell anemia, Hunt- ington disease, cystic fibrosis, Lou Gehrig disease, and Tourette syndrome, whereas most are less familiar including pachyonychia congenita (PC). Many rare diseases have patient populations of fewer than a hundred (http:// www.fda.gov/fdac/features/2003/603_orphan.html). Although the incidence of an individual rare disease is small, cumulatively the 6,000 known rare diseases affect 25 million Americans or nearly 10% of the US population (Ra- dos, 2003). The majority of rare diseases, including PC, have a genetic origin component, have little or no treatment options, and are not rigorously studied. The number of PC patients worldwide is currently unknown, but is likely to be on the order of 1,000–10,000 patients (Sancy Leachman, personal communication; see also http://www.emedicine. com/derm/topic812.htm). Costs of Developing Therapeutics According to a widely accepted study performed by DiMasi et al (2003), the estimated average out-of-pocket cost for bringing a new drug to market is $403 million (year 2000, US dollars). When investment and capitalization costs are in- cluded this estimate rises to $802 million. A more recent study by Bain & Co. put the costs at $1.7 billion (Gilbert et al, 2003; Mullin, 2003). Regardless of the actual costs, these impressively large numbers, coupled with the obser- vation that only 21% of drugs that enter phase I clinical trials make it to the marketplace, underscore the difficulty of en- ticing the investment community to fund research for rare diseases for which recovery of development costs seems unlikely. In a marketplace where the business model has historically been on blockbuster drugs that generate over a billion dollars in annual sales, there has been reluctance among large pharmaceutical companies and the investment community to pursue small market therapeutics. Resources Available to Stimulate Rare Disease Research Perhaps the most important governmental act that stimu- lated research and development of rare disease therapeu- tics was passage of the 1983 Orphan Drug Act. This program provided financial incentives to develop treatments for rare diseases, including a guarantee of a 7-y period of market exclusivity, tax credits for clinical research, and waiver provisions for license fees. The Orphan Drug Act has been successful in stimulating development of rare disease drugs (Fig 1) by small and medium-sized biotechnology companies—more than 200 drugs and biological products for the treatment of rare diseases have been brought to market since its passage in 1983. In contrast, in the 10 y prior to its passage, fewer than ten rare disease products came to the market (Rohde, 2000; Lichtenberg and Waldfo- gel, 2003; see also http://www.fda.gov/orphan). European Abbreviations: FDA, Food and Drug Administration; NIH, National Institutes of Health; ORD, Office of Rare Diseases; PC, pachyo- nychia congenita Copyright r 2005 by The Society for Investigative Dermatology, Inc. 62

Upload: roger-l

Post on 15-Jul-2016

212 views

Category:

Documents


0 download

TRANSCRIPT

Challenges in Developing Therapies for Rare Diseases IncludingPachyonychia Congenita

Roger L. KasparTransderm and SomaGenics, Inc., Santa Cruz, California, USA

The ability to attract sufficient resources to effectively develop therapeutics for rare diseases is a daunting task.

This review summarizes existing resources for rare diseases and discusses some of the challenges and strategies

associated with developing therapies for small patient populations with an emphasis on pachyonychia congenita.

Key words: orphan disease/pachyonychia congenita/rare diseaseJ Investig Dermatol Symp Proc 10:62 –66, 2005

A rare (orphan) disease is defined under the United StatesOrphan Drug Act amendment (Orphan Drug Act, P.L. 97–414(1983); Health Promotion and Disease Prevention Amend-ments, P.L. 98–551 (1984)) as a disorder that generally af-fects less than 200,000 individuals ( � 0.07% of the USpopulation). The definition is extended to disorders that af-fect greater numbers of individuals for which drug devel-opment is not likely, due to the expectation that the sales ofsuch a drug would not be sufficient to recover developmentcosts. Similar rare disease definitions are used by othercountries including the European Community, Japan,Singapore and Australia (see the websites, http://www.rare-cancer.org/rare-diseases.html and http://www.europarl.eu.int/stoa/publi/167780/chap5_en.htm).

Approximately 6,000 rare diseases have been identifiedand a list is maintained by the Office of Rare Diseases (ORD)at the National Institutes of Health (NIH) (http://ord.aspensys.com/asp/diseases/diseases.asp). Some of the listed rarediseases are well-known, such as sickle cell anemia, Hunt-ington disease, cystic fibrosis, Lou Gehrig disease, andTourette syndrome, whereas most are less familiar includingpachyonychia congenita (PC). Many rare diseases havepatient populations of fewer than a hundred (http://www.fda.gov/fdac/features/2003/603_orphan.html).

Although the incidence of an individual rare disease issmall, cumulatively the 6,000 known rare diseases affect 25million Americans or nearly 10% of the US population (Ra-dos, 2003). The majority of rare diseases, including PC,have a genetic origin component, have little or no treatmentoptions, and are not rigorously studied. The number of PCpatients worldwide is currently unknown, but is likely to beon the order of 1,000–10,000 patients (Sancy Leachman,personal communication; see also http://www.emedicine.com/derm/topic812.htm).

Costs of Developing Therapeutics

According to a widely accepted study performed by DiMasiet al (2003), the estimated average out-of-pocket cost forbringing a new drug to market is $403 million (year 2000, USdollars). When investment and capitalization costs are in-cluded this estimate rises to $802 million. A more recentstudy by Bain & Co. put the costs at $1.7 billion (Gilbertet al, 2003; Mullin, 2003). Regardless of the actual costs,these impressively large numbers, coupled with the obser-vation that only 21% of drugs that enter phase I clinical trialsmake it to the marketplace, underscore the difficulty of en-ticing the investment community to fund research for rarediseases for which recovery of development costs seemsunlikely. In a marketplace where the business model hashistorically been on blockbuster drugs that generate over abillion dollars in annual sales, there has been reluctanceamong large pharmaceutical companies and the investmentcommunity to pursue small market therapeutics.

Resources Available to Stimulate RareDisease Research

Perhaps the most important governmental act that stimu-lated research and development of rare disease therapeu-tics was passage of the 1983 Orphan Drug Act. Thisprogram provided financial incentives to develop treatmentsfor rare diseases, including a guarantee of a 7-y period ofmarket exclusivity, tax credits for clinical research, andwaiver provisions for license fees. The Orphan Drug Act hasbeen successful in stimulating development of rare diseasedrugs (Fig 1) by small and medium-sized biotechnologycompanies—more than 200 drugs and biological productsfor the treatment of rare diseases have been brought tomarket since its passage in 1983. In contrast, in the 10 yprior to its passage, fewer than ten rare disease productscame to the market (Rohde, 2000; Lichtenberg and Waldfo-gel, 2003; see also http://www.fda.gov/orphan). European

Abbreviations: FDA, Food and Drug Administration; NIH, NationalInstitutes of Health; ORD, Office of Rare Diseases; PC, pachyo-nychia congenita

Copyright r 2005 by The Society for Investigative Dermatology, Inc.

62

Orphan Medicinal Drug Product legislation similarly pro-vides financial incentives (Milne et al, 2001).

Recognizing the need to address the 25 million Ameri-cans afflicted with a rare disease, the US Congress es-tablished The ORD in 1993 to promote research andcollaborative efforts. H.R. 4014 (2002) gave statutory sta-tus to the ORD at the NIH. The ORD provides information onrare diseases, and links investigators with research subjectsand patients, and supports rare disease research (Rados,2003). In addition, the ORD funds research into rare dis-eases directly or in combination with other NIH Institutes.Furthermore, in 2003 a Rare Diseases Clinical ResearchNetwork was established, which includes seven Rare Dis-eases Clinical Research Centers spread throughout theUnited States (see website: http://rarediseases.info.nih.gov/html/resources/extr_res.html). This Network maintains adatabase of clinical trials for rare diseases and refers raredisease patients to appropriate medical care.

Other organizations have been formed to facilitate infor-mation flow and research and development of rare diseasetherapeutics. The National Organization of Rare Diseases(NORD) is a federation of approximately 125 voluntaryhealth organizations and over 60,000 patients (http://www.rarediseases.org/). The International Rare DiseaseSupport Network (IRDSN) offers support groups for over1200 diseases (http://www.raredisorders.com/). OrphaNetprovides information on rare diseases (http://www.orpha.net/consor/cgi-bin/home.php?Lng=GB). The EuropeanRare Disease Therapeutic Initiative (ERDITI, http://www.erditi.org/) is a coalition of patient organizations fosteringinteractions between academic institutions working on rarediseases and the pharmaceutical industry to bring newtherapeutics to the marketplace. Similarly, the Office of Or-phan Products Development (OOPD, http://www.fda.gov/orphan/progovw.htm) at the US Food and Drug Adminis-tration (FDA) and Public Health Programme (formerlyThe EU Programme on Rare Diseases, http://europa.eu.int/comm/health/ph_overview/previous_programme/rare_diseases/rarediseases_en.htm) facilitate orphan drug de-velopment through guidance and financial assistance (seeFig 2). The National Center For Study of Orphan Disease(CSOD, http://www.csod.us/) bridges various organizationsto facilitate rare disease therapeutic development.

Justification of Research Dollars forRare Diseases

Given the current high cost of bringing a new drug to market(see above), the difficult question arises regarding at whatlevel it becomes financially feasible to fund research thatbenefits a relatively small group of patients with a given raredisease. If the costs of developing a therapeutic to treat adisease that afflicts 10 million people is approximately thesame as a distinct therapeutic that would treat 100,000 (or100), how can the high cost of developing a therapeutic fora rare disease be justified?

A study by Love and Palmedo (Consumer Project onTechnology, (see website: http://www.cptech.org/ip/health/orphan/irsdata9798.html) challenges the reported high de-velopment costs associated with rare disease drug discov-ery. Taking into account the amount US taxpayers receivedas tax credits (50% credit allowed) for orphan disease clin-ical development ($141 million for 1997 and 1998, the mostrecent data available), they calculated the cost for clinicaldevelopment, testing and marketing of the 36 orphanproducts approved during the same time period to be$7.9 million per orphan product ($283 million/36 products)before tax, and $3.9 million with the benefits of the orphandrug tax credit.

A number of biotechnology companies have taken ad-vantage of the Orphan Drug Act financial incentives to spe-cifically develop and market therapeutics for the raredisease market. Several companies have profitably target-ed this niche market. Approximately half of Genzyme’s 2003revenue came from $740 million in sales of Cerezyme(Genzyme, Cambridge, MA). This is an enzyme replacementtherapy for Gaucher disease, a potentially deadly genetic

Figure 1Orphan drug approval. Cumulative number of orphan drugs approvedfrom 1979 through 1998 as compared to cumulative number of totaldrugs approved. The Orphan Drug Act was passed in 1983. Graphreproduced with permission from Lichtenberg (Lichtenberg andWaldfogel, 2003).

Figure2Flowchart of milestones leading to approval of pachyonychia con-genita (PC) therapeutic. New orphan drug development process. Ap-plication for orphan drug status (for therapeutics being developed fordiseases afflicting fewer than 200,000 patients) can be made at anytime prior to new drug application (NDA) application. The Food andDrug Administration (FDA) Office of Orphan Products Development(OOPD) helps facilitate orphan disease product development and en-courages interaction between sponsor, FDA and OOPD at variousstages of the process including prior to application for orphan drugstatus. Figure was modified (with permission) from the FDA website(http://www.fda.gov/cder/handbook/develop.htm).

DEVELOPING A PC THERAPEUTIC 6310 : 1 OCTOBER 2005

disorder that causes anemia and enlarged organs (http://www.genzyme.com). Gaucher disease affects less than10,000 people worldwide and about 40% are treated withCerezyme. Although the 7 y marketing exclusivity of Cere-zyme has long since ended, the drug remains entrenched inthe marketplace and highly profitable. Much of the remain-der of Genzyme’s sales comes from other therapeutics withcurrent orphan drug status including Fabrazyme (for Fabrydisease), Aldurazyme (for Mucopolysaccharidosis I), andThyrogen (for thyroid cancer). Other companies includingTranskaryotic Therapies (Cambridge, MA), Oxford Glyco-Sciences (Brussels, Belgium), and Orphan Medical (Minne-tonka, MN) have also taken a similar approach to seek nichemarkets for specific rare diseases, taking advantage ofthe Orphan Drug Act. Many current blockbuster drugs(sales over $1 billion per year), including Amgen’s Epogenand Neupogen, were originally introduced as orphan drugproducts and were later extended to larger markets.

Societal Benefits of Rare Disease Research

Why treat rare diseases, particularly diseases in which verysmall numbers of patients are involved? There are severalanswers to this question, not the least of which is that theseare real people with families and employers that are alsoaffected by the loss of quality of life and productivity. Fur-thermore, science is replete with examples of esoteric re-search that has led to unintended discoveries that benefitsociety at large. Basic research is funded with the expec-tation that investigation in one area will have benefits inmany related as well as unrelated and unexpected areas.The study of rare diseases has often yielded great amountsof information, completely out of proportion with thenumber of patients suffering from the disorder. William Ha-rvey, the English physician and discoverer of blood circu-lation, stated in a letter dated 1657, ‘‘Nature is no whereaccustomed more openly to display her secret mysteriesthan in cases where she shows traces of her workings apartfrom the beaten path; nor is there any better way to ad-vance the proper practice of medicine than to give ourminds to the discovery of the usual law of nature, by carefulinvestigation of cases of rarer forms of disease. For it hasbeen found in almost all things that what they contain ofuseful or applicable, is hardly perceived unless we are de-prived of them, or they become deranged in some way(Willis, 1847; Zelzer and Olsen, 2003).’’

The majority of rare diseases are due to genetic muta-tions. In many cases, these mutations affect a single geneand cause perturbations in a metabolic pathway. This raredisease datasource has been a rich resource from which agreat deal has been learned about normal human metab-olism, since the study of aberrant metabolism (i.e., blockageof a single step in a metabolic pathway due to the absenceof a functional gene) teaches a great deal regarding hownormal pathways function. These ‘‘natural’’ experiments aresimilar to carefully controlled knockout animal experimentsin which the function of single genes are analyzed and oftengive major insights into metabolic pathways.

There are numerous examples in which rare disease re-search has led to insights into more general disease proc-

esses. For instance, it would have been difficult to predictthat study of kuru, a mental illness in a New Guinea tribe,one symptom of which is uncontrollable laughter, wouldlead to the discovery of a new class of contagion and resultin a Nobel prize for Stanley Prusiner (Prusiner, 1984). Thediscovery that the disease-causing prions were passedalong by consumption of the brains of deceased relativesallowed the discovery of the prion cause of other rarediseases including Creutzfeldt–Jakob and Gertsmann–Straussler–Scheinker (GSS) syndrome, as well as recogniz-ing that bovine spongiform encephalopathy (BSE), wascaused by ingestion of contaminated beef. This pioneeringwork likely resulted in an epidemic being averted and thou-sands of lives spared. Another example is a-1-antitrypsindeficiency, a rare genetic emphysema lung disease thatdevelops 10–30 y before the occurrence of the more com-mon form found in smokers. The earlier onset has allowedresearchers to study the disorder in the absence of com-pounding factors due to smoking and aging, leading to in-sights into emphysema (Rados, 2003).

Leveraging Existing Resources

PC is an excellent example of a rare disease that couldserve as a model for a multitude of other skin disorders and/or autosomal dominant disorders. As discussed above andin other reports in this issue, the specific mutations in ker-atin genes K6a/b, K16 and K17 responsible for PC havebeen identified (Munro, 2001; Terrinoni et al, 2001; Smith,2003). Furthermore, the skin cell type involved (keratinocyte)is known and is readily accessible. Therefore, this disordercould serve as a paradigm for many other keratinocyte skindisorders, including psoriasis and epidermolysis bullosa(Porter and Lane, 2003; Sawamura et al, 2003). Technolo-gies including siRNA, ribozymes, and antisense have beenshown to specifically block expression of target genes inmany systems (see Lewin et al, this issue) including kera-tinocytes (Mehta et al, 2000; Arts et al, 2003; Barbieri et al,2003; Seo et al, 2004). The main remaining hurdle for clinicalapplication is efficient delivery to the appropriate cells. De-velopment of a skin delivery system in an ‘‘easy to workwith’’ system would be a boon to treatment of other skindisorders. PC may represent one of the ‘‘most straightfor-ward’’ genetic disorders for treatment due to the accessi-bility of the skin. Furthermore, complete blockage orremoval of a mutant keratin gene may not be necessaryas a partial reduction may be sufficient to give a therapeuticeffect (see discussion of K10/K14 in Chen and Roop, thisissue). Unlike psoriasis, which involves multiple cell typesincluding keratinocytes and immune cells (Kirby andGriffiths, 2002; Barry and Kirby, 2004), PC appears to belimited to keratinocytes. Finally, the difficulty of discrimina-tion between mutant and wild-type genes, likely to be nec-essary for most genetic diseases, does not appear to be arequisite for an effective PC treatment. Reduction of boththe wild-type and mutant keratin gene products is likely togive a therapeutic effect as other ‘‘redundant’’ keratin pairswill likely compensate for the missing mutant keratin genepair (see Lewin et al, in this issue).

64 KASPAR JID SYMPOSIUM PROCEEDINGS

Bringing Together Existing Expertise andResources for Rare Disease Therapeutic

Development

The Pachyonychia Congenita Public Charity was formedwith the aim to develop an effective therapeutic for the rel-atively small number of patients suffering from PC (http://www.pachyonychia.org/index.html). To this end, a consor-tium of investigators was brought together with medical andscientific expertise in various areas related to PC and tech-nology that may be useful in developing PC therapeutics.The Public Charity has provided seed funding to allow tar-geted research that should facilitate progress towards PCtherapeutic development (see Introductory PC article in thisissue). The collaborative nature of the consortium is essen-tial to prevent wasteful redundant research and allow effi-cient use of limited resources. Furthermore, the breadth ofthe expertise of the consortium membership will allow con-tinual monitoring of scientific and medical progress ofrelated skin disorders, ideally taking advantage of andbuilding on breakthroughs including new skin deliverysystems.

In addition to the Pachyonychia Congenita Public Char-ity, other private organizations and philanthropic individuals,public resources are available for rare diseases researchand clinical development and testing including the NIHR21 Exploratory and Development (http://grants1.nih.gov/grants/guide/pa-files/PA-03-171.html) and U54 (http://grants.nih.gov/grants/guide/rfa-files/RFA-RR-03-008.html) grantmechanisms. Furthermore, the FDA administers a programto fund clinical trials for drugs that have achieved orphandrug status (see website: http://www.fda.gov/orphan/grants/2004RFA.htm).

Other Considerations

As discussed in other reports in this PC JID edition, the idealsolution for a permanent cure for PC would be a gene ther-apy replacement procedure in which the defective PC genewould be replaced with a corrected version that would beregulated in identical fashion to the wild-type gene. This‘‘gold standard’’ gene replacement therapy has not beenapproved for any disorder to date and will likely be initiallyapplied to acute, life threatening, ‘‘no alternative’’ diseases.The amount of development and testing required, as well asits non-life threatening nature, makes PC an unlikely earlycandidate for gene therapy. On the other hand, PC is astraightforward dominant-negative genetic skin disorder inwhich the responsible gene mutations have been identified.Animal experiments suggest that therapeutic benefits mayaccrue from simple downregulation of the mutant gene.Surprisingly, downregulation of the wild-type K6a genedoes not result in apparent skin structural defects, as otherrelated keratin gene pairs appear to compensate for thedeficiency (Wojcik et al, 2000; Wojcik et al, 2001), see alsoChen and Roop, in this issue. Therefore, as replacementgene therapy technology improves and safer human genedelivery vehicles are developed, PC will be an ideal genetherapy candidate. In the short run, however, non-viral de-livery of potent specific keratin inhibitors, preferably deliv-

ered topically to avoid systemic side effects, appears to bethe most promising approach from a practical, regulatory,ethical and economical standpoint.

Conclusions

Personalized molecular medicine: the end of rare dis-eases? With the advent of new technologies including mi-croarray chips that can detect steady-state levels of tissuemRNA including single nucleotide mutations, the era ofpersonalized molecular medicine appears imminent (Jain,2002, 2004). Powerful technologies continue to be devel-oped that can specifically target and block expression ofspecific genes (the latest being RNA interference or siRNA(Dave and Pomerantz, 2003; Alisky and Davidson, 2004;Caplen, 2004; see also Lewin et al, in this issue). Asprogress continues in the arena of specific and safe deliveryvectors for correcting genes or delivering siRNA or othergene-specific inhibitors directly, one can envision a scenarioin which diagnosis of overexpressed genes involved in rarediseases is readily translated into designer molecular med-icines that specifically target these problem genes. If thedelivery issue can be solved, theoretically any gene can betargeted and its protein product blocked or reduced, pro-viding a therapeutic effect. Specifically targeting problem-atic single nucleotide mutations in disease-causing geneswill likely prove more difficult and may require improvedtechnology. Some diseases, such as PC, however, may notsuffer from this discrimination difficulty as concomitant re-duction of the wild-type gene is unlikely to affect the ther-apeutic outcome as other keratin gene pairs maycompensate as a result of redundancy in the system (Wonget al, 2000; Wojcik et al, 2001; Wong and Coulombe, 2003).This advantage, coupled with relatively easy access to thediseased cells (skin keratinocytes), makes PC an excellentcandidate disorder for treatment with specific and robustnucleic acid therapeutics such as siRNA.

DOI: 10.1111/j.1087-0024.2005.10208.x

Manuscript received June 9, 2005; revised June 27, 2005; accepted forpublication June 28, 2005

Address correspondence to: Roger L. Kaspar, Transderm, 2161Delaware Ave., Santa Cruz, CA 95060, USA. Emails: [email protected]

References

Alisky JM, Davidson BL: Towards therapy using RNA interference. Am J

Pharmacogenomics 4:45–51, 2004

Arts GJ, Langemeijer E, Tissingh R, et al: Adenoviral vectors expressing siRNAs

for discovery and validation of gene function. Genome Res 13:2325–

2332, 2003

Barbieri CE, Barton CE, Pietenpol JA: Delta Np63 alpha expression is regulated

by the phosphoinositide 3-kinase pathway. J Biol Chem 278:51408–

51414, 2003

Barry J, Kirby B: Novel biologic therapies for psoriasis. Expert Opin Biol Ther

4:975–987, 2004

Caplen NJ: Gene therapy progress and prospects. Downregulating gene expres-

sion: The impact of RNA interference. Gene Ther 11:1241–1248, 2004

Chen J, Roop DR: Mouse models in pre-clinical studies for pachyonychia

congenita. J Investig Dermatol 10:37–46, 2005

DEVELOPING A PC THERAPEUTIC 6510 : 1 OCTOBER 2005

Dave RS, Pomerantz RJ: RNA interference: On the road to an alternate thera-

peutic strategy!. Rev Med Virol 13:373–385, 2003

DiMasi JA, Hansen RW, Grabowski HG: The price of innovation: New estimates of

drug development costs. J Health Econ 22:151–185, 2003

Gilbert J, Henske P, Singh A: Rebuilding big pharma’s business model. In Vivo:

Business Med Rep 21:73, 2003

Jain KK: Personalized medicine. Curr Opin Mol Ther 4:548–558, 2002

Jain KK: Applications of biochips: From diagnostics to personalized medicine.

Curr Opin Drug Discov Devel 7:285–289, 2004

Kirby B, Griffiths CE: Novel immune-based therapies for psoriasis. Br J Dermatol

146:546–551, 2002

Lewin AS, Glazer PM, Milstone LM: Gene therapy for autosomal dominant

disorders of keratin. J Investig Dermatol 10:47–61, 2005

Mehta RC, Stecker KK, Cooper SR, et al: Intercellular adhesion molecule-1 sup-

pression in skin by topical delivery of anti-sense oligonucleotides. J Invest

Dermatol 115:805–812, 2000

Mullin R: Drug development costs about $1.7 billion. Chem Eng News 81:8, 2003

Munro CS: Pachyonychia congenita: Mutations and clinical presentations. Br J

Dermatol 144:929–930, 2001

Porter RM, Lane EB: Phenotypes, genotypes and their contribution to under-

standing keratin function. Trends Genet 19:278–285, 2003

Prusiner SB: Prions: Novel infectious pathogens. Adv Virus Res 29:1–56, 1984

Rados C: Orphan products: Hope for people with rare diseases. FDA Consumer

Mag 37:10–15, 2003

Rohde DD: The Orphan Drug Act: An engine of innovation? At what cost? Food

Drug Law J 55:125–143, 2000

Sawamura D, McMillan JR, Akiyama M, Shimizu H: Epidermolysis bullosa: Di-

rections for future research and new challenges for treatment. Arch De-

rmatol Res 295 (Suppl 1):S34–S42, 2003

Seo M, Cho CH, Lee YI, et al: Cdc42-dependent mediation of UV-induced p38

activation by G protein betagamma subunits. J Biol Chem 279:17366–

17375, 2004

Smith F: The molecular genetics of keratin disorders. Am J Clin Dermatol 4:347–

364, 2003

Terrinoni A, Smith FJ, Didona B, et al: Novel and recurrent mutations in the genes

encoding keratins K6a, K16 and K17 in 13 cases of pachyonychia con-

genita. J Invest Dermatol 117:1391–1396, 2001

Willis R: The Works of William Harvey. London: Sydenham Society, 1847

Wojcik SM, Bundman DS, Roop DR: Delayed wound healing in keratin 6a

knockout mice. Mol Cell Biol 20:5248–5255, 2000

Wojcik SM, Longley MA, Roop DR: Discovery of a novel murine keratin 6 (K6)

isoform explains the absence of hair and nail defects in mice deficient for

K6a and K6b. J Cell Biol 154:619–630, 2001

Wong P, Colucci-Guyon E, Takahashi K, Gu C, Babinet C, Coulombe PA: Intro-

ducing a null mutation in the mouse K6alpha and K6beta genes reveals

their essential structural role in the oral mucosa. J Cell Biol 150:921–928,

2000

Wong P, Coulombe PA: Loss of keratin 6 (K6) proteins reveals a function for

intermediate filaments during wound repair. J Cell Biol 163:327–37,

2003

Zelzer E, Olsen BR: The genetic basis for skeletal diseases. Nature 423:343–348,

2003

66 KASPAR JID SYMPOSIUM PROCEEDINGS