Molecular Understanding and Modern Application of Traditional Medicines: Triumphs and Trials

生物 计算生物学 工程伦理学 生物信息学 工程类
作者
Timothy W. Corson,Craig M. Crews
出处
期刊:Cell [Elsevier]
卷期号:130 (5): 769-774 被引量:488
标识
DOI:10.1016/j.cell.2007.08.021
摘要

Traditional medicines provide fertile ground for modern drug development, but first they must pass along a pathway of discovery, isolation, and mechanistic studies before eventual deployment in the clinic. Here, we highlight the challenges along this route, focusing on the compounds artemisinin, triptolide, celastrol, capsaicin, and curcumin. Traditional medicines provide fertile ground for modern drug development, but first they must pass along a pathway of discovery, isolation, and mechanistic studies before eventual deployment in the clinic. Here, we highlight the challenges along this route, focusing on the compounds artemisinin, triptolide, celastrol, capsaicin, and curcumin. Traditional medicines continue to provide front-line pharmacotherapy for many millions of people worldwide. Although their application is often viewed with skepticism by the Western medical establishment, medicinal extracts used in ancient medical traditions such as Ayurveda on the Indian subcontinent and traditional Chinese medicine (TCM) are a rich source of therapeutic leads for the pharmaceutical industry. The transformation of traditional medicines into modern drugs has its origins in the archetypal examples of the antimalarial quinine and the antipyretic analgesic aspirin. The alkaloid quinine was isolated in 1820 from the bark of several species of Cinchona, thought to have been used by Peruvian Indians to suppress shivering and used since the 17th century in the treatment of malarial fevers (Greenwood, 1992Greenwood D. J. Antimicrob. Chemother. 1992; 30: 417-427Crossref PubMed Scopus (56) Google Scholar). Similarly, aspirin was derived from salicylic acid in the bark of the willow tree (Salix species), used traditionally to treat fever and inflammation in many cultures worldwide for at least four millennia (Mahdi et al., 2006Mahdi J.G. Mahdi A.J. Mahdi A.J. Bowen I.D. Cell Prolif. 2006; 39: 147-155Crossref PubMed Scopus (117) Google Scholar). The successes of these two early “blockbuster” drugs set the stage for ongoing drug discovery efforts from traditional medicines. Compounds derived from medicinal extracts are appealing for several reasons (Schmidt et al., 2007Schmidt B.M. Ribnicky D.M. Lipsky P.E. Raskin I. Nat. Chem. Biol. 2007; 3: 360-366Crossref PubMed Scopus (243) Google Scholar). They are often stereochemically complex, multi- or macrocyclic molecules with limited likelihood of prior chemical synthesis, and they tend to have interesting biological properties. But perhaps most importantly, parent extracts have been “clinically” tested in their traditional milieu, in some cases over millennia. Despite these advantages, the path from traditional medicine to Western pharmaceutical is fraught with challenges. Here, we discuss the challenges of each of the four steps in this pipeline (see Figure 1): Western “discovery” of a traditional medicine, isolation and/or synthesis of the active component, elucidation of the molecular mechanism, and development as a pharmaceutical. We focus on five interesting and timely examples derived from traditional medicines in varied therapeutic classes, each at a different stage in the development process, highlighting successes and roadblocks on the path to status as a Western drug. The antimalarial artemisinin (and derivatives) represents one of the greatest recent clinical success stories arising from a traditional medicine, echoing the success of quinine two centuries earlier. Artemisinin (see Figure 1) is derived from Artemisia annua L., the sweet wormwood (qinghao), a shrub first documented in TCM in 168 BCE as a hemorrhoid treatment (Liu et al., 2006Liu C. Zhao Y. Wang Y. Appl. Microbiol. Biotechnol. 2006; 72: 11-20Crossref PubMed Scopus (129) Google Scholar). Since at least the fourth century CE, it has been used in the treatment of fever attributed to malaria. This long history of use prompted Chinese researchers to seek the active antimalarial principle; artemisinin was isolated and its structure determined in the mid 1970s (Liu et al., 2006Liu C. Zhao Y. Wang Y. Appl. Microbiol. Biotechnol. 2006; 72: 11-20Crossref PubMed Scopus (129) Google Scholar). Artemisinin, an endoperoxide sesquiterpene lactone with a complex polycyclic ring structure, is modified by Fe2+ ions to structures containing carbon-centered free radicals. Given that the intracellular environment of the Plasmodium malaria parasite is rich in this ion from heme, these radicals are currently thought to be responsible for artemisinin's antimalarial activity. The classic method of cell fractionation after treatment with radiolabeled artemisinin has identified numerous cellular constituents alkylated by artemisinin (Asawamahasakda et al., 1994Asawamahasakda W. Ittarat I. Pu Y.M. Ziffer H. Meshnick S.R. Antimicrob. Agents Chemother. 1994; 38: 1854-1858Crossref PubMed Scopus (200) Google Scholar); the strongest validated target for artemisinin is PfATP6, the Plasmodium sarco-endoplasmic reticulum Ca2+ ATPase (SERCA), which is inhibited by artemisinin (Eckstein-Ludwig et al., 2003Eckstein-Ludwig U. Webb R.J. Van Goethem I.D. East J.M. Lee A.G. Kimura M. O'Neill P.M. Bray P.G. Ward S.A. Krishna S. Nature. 2003; 424: 957-961Crossref PubMed Scopus (830) Google Scholar). Clinical studies, initiated in the 1970s prior to any mechanistic insights into artemisinin function, demonstrated that artemisinin and its derivatives are powerful antimalarials. They have proved particularly effective for treating severe malaria and, in combination with traditional antimalarials, for combatting Plasmodium drug resistance. Combination therapies containing artemisinin are now considered the treatment of choice for malaria in Asia, with growing adoption in Africa (see Table 1 for information on clinical trials). Artemisinin may also have efficacy against other parasites and as an anticancer compound, possibly acting via antiangiogenic and proapoptotic mechanisms in the latter case (Efferth, 2007Efferth T. Planta Med. 2007; 73: 299-309Crossref PubMed Scopus (232) Google Scholar).Table 1Five Traditional Medicines in Clinical TrialsCompoundDiseaseClinical TrialsNumberPrincipal SponsorsArtemisininMalaria8131 charities, institutes, universities, and companies based in Australia, Austria, Belgium, Colombia, Ethiopia, France, Gambia, Germany, Ghana, Guinea-Bissau, the Netherlands, Papua New Guinea, South Africa, Sudan, Sweden, Switzerland, the United Kingdom, and the USA; and working in numerous Asian, African, and South American locationsCytomegalovirus infection1Hadassah Medical Organization, IsraelSchistosomiasis1Dafra Pharma, BelgiumTriptolide & Celastrol (T. wilfordii extract)Rheumatoid arthritis1National Institute of Arthritis and Musculoskeletal and Skin Diseases, MD, USACapsaicinChronic pain13NeurogesX, CA, USA; AlgoRx Pharmaceuticals, NJ, USAPostoperative pain5National Institute of Dental and Craniofacial Research, MD, USA; AlgoRx Pharmaceuticals, NJ, USARadiation-induced mucositis1North Central Cancer Treatment Group, MN, USAAlopecia areata1University of Minnesota, USAMorton's neuroma1AlgoRx Pharmaceuticals, NJ, USAOsteoarthritis1Khon Kaen University, ThailandInterstitial cystitis1National Institute of Diabetes and Digestive and Kidney Diseases, MD, USACurcuminColon cancer6Chao Family Comprehensive Cancer Center, CA, USA; Tel-Aviv Sourasky Medical Center, Israel; Johns Hopkins University, MD, USA; University of Michigan Comprehensive Cancer Center, USA; University of Pennsylvania, USA; University of Medicine and Dentistry, NJ, USAPancreatic cancer3Rambam Medical Center, Israel; M.D. Anderson Cancer Center, TX, USA; Tel-Aviv Sourasky Medical Center, IsraelAlzheimer's disease2John Douglas French Foundation, CA, USA; Chinese University of Hong KongChemotherapy-induced mucositis1Hadassah Medical Organization, IsraelMultiple myeloma1M.D. Anderson Cancer Center, TX, USAPsoriasis1University of Pennsylvania, USACystic fibrosis1Seer Pharmaceuticals, CT, USAIncludes registered, open, closed, terminated, and completed trials of these compounds, parent extracts, or derivatives. For details see www.clinicaltrials.gov. Data current as of August 21, 2007. Open table in a new tab Includes registered, open, closed, terminated, and completed trials of these compounds, parent extracts, or derivatives. For details see www.clinicaltrials.gov. Data current as of August 21, 2007. Despite these dramatic findings, widespread deployment of artemisinin has been hindered by production difficulties. Although a dozen synthetic routes to artemisinin have been described, all are complex and low yielding, rendering them economically unfeasible (Liu et al., 2006Liu C. Zhao Y. Wang Y. Appl. Microbiol. Biotechnol. 2006; 72: 11-20Crossref PubMed Scopus (129) Google Scholar). Synthetic chemistry has, however, offered semi-synthetic artemisinin derivatives with improved solubility (such as sodium artesunate) and stability (such as artemether) (Efferth, 2007Efferth T. Planta Med. 2007; 73: 299-309Crossref PubMed Scopus (232) Google Scholar). Even a totally synthetic trioxolane compound RBX11160 (OZ277), inspired by the trioxane endoperoxide moiety of artemisinin, has shown promise as an antimalarial (Vennerstrom et al., 2004Vennerstrom J.L. Arbe-Barnes S. Brun R. Charman S.A. Chiu F.C. Chollet J. Dong Y. Dorn A. Hunziker D. Matile H. et al.Nature. 2004; 430: 900-904Crossref PubMed Scopus (557) Google Scholar). Artemisinin for clinical use is predominantly produced naturally in A. annua plants. Despite efforts to maximize agricultural production, the artemisinin content in plant extracts varies widely due to environmental conditions: 0.01%–0.8% dry weight (Efferth, 2007Efferth T. Planta Med. 2007; 73: 299-309Crossref PubMed Scopus (232) Google Scholar). This in turn makes the drug itself expensive—particularly problematic for an antimalarial, which is needed in large quantities in many poorer countries. Cell and plantlet cultures are an appealing alternative source of this compound as they can be grown under much more closely controlled conditions than whole plants. Indeed, useful yields of the compound can be produced by feeding cultures artemisinin precursors (Liu et al., 2006Liu C. Zhao Y. Wang Y. Appl. Microbiol. Biotechnol. 2006; 72: 11-20Crossref PubMed Scopus (129) Google Scholar). An alternative approach is the genetic engineering of A. annua itself. The plant has proven genetically tractable: Several of the isoprenoid biosynthetic enzymes necessary for artemisinin production have been cloned, and A. annua can be successfully transformed with Agrobacterium tumefaciens to overexpress key biosynthetic genes (Liu et al., 2006Liu C. Zhao Y. Wang Y. Appl. Microbiol. Biotechnol. 2006; 72: 11-20Crossref PubMed Scopus (129) Google Scholar). Perhaps the most promising strategy is the use of microbes to produce artemisinin. In a triumph of genetic engineering, Ro et al. combined genetic activation of the endogenous mevalonate isoprenoid synthesis pathway with introduction of A. annua genes to produce artemisinic acid in the budding yeast Saccharomyces cerevisiae (Ro et al., 2006Ro D.K. Paradise E.M. Ouellet M. Fisher K.J. Newman K.L. Ndungu J.M. Ho K.A. Eachus R.A. Ham T.S. Kirby J. et al.Nature. 2006; 440: 940-943Crossref PubMed Scopus (1978) Google Scholar). This precursor compound, which can be readily converted to artemisinin in the laboratory, is secreted in large quantities from the yeast. Such creative strategies, leveraging the power of genetics and in vivo biochemistry, can provide a valuable counterpart to synthetic chemistry and natural sources in the production of natural product medicines. Trypterygium wilfordii Hook F., the “thunder god vine” (lei gong teng), is another TCM. This vine has been used traditionally for the treatment of arthritis and other diseases, and it is the source of several biologically active secondary metabolites (Tao and Lipsky, 2000Tao X. Lipsky P.E. Rheum. Dis. Clin. North Am. 2000; 26: 29-50Abstract Full Text Full Text PDF PubMed Scopus (240) Google Scholar). Some of its TCM uses might rely on the presence of multiple active components, and clinical studies have been performed on extracts of the plant (Table 1), rather than on a single compound (Tao and Lipsky, 2000Tao X. Lipsky P.E. Rheum. Dis. Clin. North Am. 2000; 26: 29-50Abstract Full Text Full Text PDF PubMed Scopus (240) Google Scholar). However, substantial work has focused on two major bioactive constituent compounds: triptolide and celastrol (Figure 1). Triptolide is a diterpenoid epoxide with a staggering variety of documented cellular effects. Along with anti-inflammatory activity, it shows anticancer, immunosuppressive, and antifertility effects (Qiu and Kao, 2003Qiu D. Kao P.N. Drugs R D. 2003; 4: 1-18Crossref PubMed Scopus (274) Google Scholar). It was isolated in 1972, and several synthetic routes have been described since then (Yang et al., 1998Yang D. Ye X.-Y. Xu M. Pang K.-W. Zou N. Letcher R.M. J. Org. Chem. 1998; 63: 6446-6447Crossref Scopus (31) Google Scholar and references therein). Like artemisinin, however, triptolide is currently derived from its plant of origin with low yield: 6–16 ng/g in one study (Brinker and Raskin, 2005Brinker A.M. Raskin I. J. Chromatogr. A. 2005; 1070: 65-70Crossref PubMed Scopus (48) Google Scholar). Little work has been done to investigate biotechnological routes to triptolide production, which are important to reduce reliance on the natural source. Moreover, continued development of derivatives of triptolide such as the succinyl sodium salt PG490-88 will be valuable to improving the solubility and side-effect profile of this compound (Tao and Lipsky, 2000Tao X. Lipsky P.E. Rheum. Dis. Clin. North Am. 2000; 26: 29-50Abstract Full Text Full Text PDF PubMed Scopus (240) Google Scholar). Determination of triptolide's cellular target has proven to be an even greater challenge. This is not unusual: Many a promising therapeutic natural product has faltered when no clear-cut mechanism of action could be identified. Although progression into the clinic without such knowledge is possible, as was the case with artemisinin, a solid knowledge of molecular mechanism (ideally at the structural, not just the molecular, level) allows medicinal chemists to perform rational derivatization to improve affinity, specificity, pharmacokinetics, and stability. Knowledge of mechanism can also potentially lead to more specific clinical trials and, in cases like triptolide, completely new insights. A large body of work describes triptolide's inhibitory effects on transcription mediated through NF-κB and NFAT (Qiu and Kao, 2003Qiu D. Kao P.N. Drugs R D. 2003; 4: 1-18Crossref PubMed Scopus (274) Google Scholar), but until recently, direct cellular targets were elusive. Nonetheless, careful cell fractionation with [3H]-triptolide enabled identification of the Ca2+ channel polycystin-2 (encoded by the PKD2 gene) as a possible triptolide-binding protein (others also likely exist) (Leuenroth et al., 2007Leuenroth S.J. Okuhara D. Shotwell J.D. Markowitz G.S. Yu Z. Somlo S. Crews C.M. Proc. Natl. Acad. Sci. USA. 2007; 104: 4389-4394Crossref PubMed Scopus (200) Google Scholar). PKD2 or the gene encoding its activator, PKD1, causes polycystic kidney disease (PKD) when mutated because entry of Ca2+ ions is essential for growth arrest of epithelial cells forming the kidney tubule. Because triptolide activates opening of the polycystin-2 channel, it could potentially complement loss of PKD1. This is the case in a mouse model of polycystic kidney disease in which the mice lack Pkd1 (Leuenroth et al., 2007Leuenroth S.J. Okuhara D. Shotwell J.D. Markowitz G.S. Yu Z. Somlo S. Crews C.M. Proc. Natl. Acad. Sci. USA. 2007; 104: 4389-4394Crossref PubMed Scopus (200) Google Scholar). Thus, this calcium-dependent activity of triptolide, which is unrelated to its transcriptional repression activity (Leuenroth and Crews, 2005Leuenroth S.J. Crews C.M. Chem. Biol. 2005; 12: 1259-1268Abstract Full Text Full Text PDF PubMed Scopus (43) Google Scholar), opens a new therapeutic avenue for pursuing triptolide, in addition to its effects on the immune and reproductive systems and in cancer. Highlighting the complexity of plant extracts, the pentacyclic triterpene celastrol (Figure 1) is structurally a very different component of T. wilfordii with a divergent therapeutic profile. Celastrol (also known as tripterine) is extracted in small quantities from T. wilfordii or other members of the Celastraceae (bittersweet) family. To our knowledge, no total synthesis or alternative production routes have been reported. Although not yet tested as a single agent in humans (Table 1), celastrol has shown promise as an anti-inflammatory compound in animal models of arthritis, lupus, amyotrophic lateral sclerosis, and Alzheimer's disease (Sethi et al., 2007Sethi G. Ahn K.S. Pandey M.K. Aggarwal B.B. Blood. 2007; 109: 2727-2735Crossref PubMed Scopus (256) Google Scholar and references therein). It also has antiproliferative effects against numerous cancer cell lines. Several molecular mechanisms have been identified for these effects, including gene expression modulation likely mediated through inhibition of NF-κB via TAK1 and IκBα kinase (Sethi et al., 2007Sethi G. Ahn K.S. Pandey M.K. Aggarwal B.B. Blood. 2007; 109: 2727-2735Crossref PubMed Scopus (256) Google Scholar and references therein), proteasome inhibition, topoisomerase II inhibition, and heat shock response activation (Hieronymus et al., 2006Hieronymus H. Lamb J. Ross K.N. Peng X.P. Clement C. Rodina A. Nieto M. Du J. Stegmaier K. Raj S.M. et al.Cancer Cell. 2006; 10: 321-330Abstract Full Text Full Text PDF PubMed Scopus (416) Google Scholar and references therein). Nonetheless, direct targets remain elusive. As celastrol and triptolide move into human studies, it will be vital not only to better understand their mechanisms of action but also to investigate any potential synergistic effects of the two compounds, both at the cellular and organismal levels. Used worldwide, the alkaloid capsaicin is the main cause of the “hot” sensation associated with chili peppers, members of the genus Capsicum. Beyond their widespread use as a spice, chili peppers were used in the Americas by the Aztecs and Tarahumara Indians as a remedy for coughs and bronchitis. Similar uses plus anti-inflammatory and gastrointestinal applications were adopted in India after the Portuguese imported chili peppers in the late 15th century. In Africa, they are traditionally used internally and externally as antiseptics (Dasgupta and Fowler, 1997Dasgupta P. Fowler C.J. Br. J. Urol. 1997; 80: 845-852Crossref PubMed Google Scholar). However, modern usage of capsaicin is focused on the treatment of various types of pain (see below) and also in the treatment of detrusor hyper-reflexia, a form of urinary incontinence (Dasgupta and Fowler, 1997Dasgupta P. Fowler C.J. Br. J. Urol. 1997; 80: 845-852Crossref PubMed Google Scholar). High-dose oral capsaicin also has anticancer properties in some animal model studies but seems to be a cancer promoter in others. Compared with artemisinin, triptolide, and celastrol, capsaicin is chemically quite simple (Figure 1). It was purified and named in the 19th century and first synthesized in the 1920s (Dasgupta and Fowler, 1997Dasgupta P. Fowler C.J. Br. J. Urol. 1997; 80: 845-852Crossref PubMed Google Scholar). But the widespread cultivation of Capsicum makes synthesis unnecessary, as large quantities of capsaicin can easily be extracted from readily available peppers. The mechanism of capsaicin in pain induction has been the topic of much neurophysiological research (Cortright et al., 2007Cortright D.N. Krause J.E. Broom D.C. Biochim. Biophys. Acta. 2007; 1772: 978-988Crossref PubMed Scopus (81) Google Scholar). Capsaicin, along with thermal heat, directly activates nociceptors in the skin, the sensory neurons responsible for the sensation of pain, with the subsequent release of the neurotransmitter substance P. Capsaicin's therapeutic effect on pain is due to the desensitization and eventual destruction of nociceptors following repeated capsaicin exposure. In a classic example of expression cloning, Caterina et al. identified the capsaicin receptor (Caterina et al., 1997Caterina M.J. Schumacher M.A. Tominaga M. Rosen T.A. Levine J.D. Julius D. Nature. 1997; 389: 816-824Crossref PubMed Scopus (6541) Google Scholar). Capsaicin was known to cause Ca2+ ion influx into nociceptors, so these authors transfected a nociceptor cDNA library into nonexcitable HEK293 cells and screened for capsaicin-dependent Ca2+ ion influx. The receptor they cloned, now known as TRPV1, is a Ca2+ ion channel that also responds to, and integrates, signals from piperine (the irritant in black pepper), protons, and other noxious stimuli (Caterina et al., 1997Caterina M.J. Schumacher M.A. Tominaga M. Rosen T.A. Levine J.D. Julius D. Nature. 1997; 389: 816-824Crossref PubMed Scopus (6541) Google Scholar). The cloning of TRPV1 kick started the field of pain receptor pharmacology. Numerous pharmaceutical companies are developing both TRPV1 antagonists (to block nociception directly) and agonists (to desensitize nociceptors, as with capsaicin) (Immke and Gavva, 2006Immke D.C. Gavva N.R. Semin. Cell Dev. Biol. 2006; 17: 582-591Crossref PubMed Scopus (136) Google Scholar). Resiniferatoxin, another traditional medicine from the latex of Euphorbia resinifera, is one such agonist with higher potency than capsaicin (Immke and Gavva, 2006Immke D.C. Gavva N.R. Semin. Cell Dev. Biol. 2006; 17: 582-591Crossref PubMed Scopus (136) Google Scholar). Efforts continue to create TRPV1 agonists with better skin permeation and lacking the distinctive side effect of a burning sensation on application. Capsaicin itself has been used clinically with moderate success as a topical treatment for the pain of rheumatoid and osteoarthritis, psoriasis, diabetic neuropathy, and postherpetic neuralgia (Table 1), but herein lies the particular challenge with this molecule: The chronic pain disorders are notoriously idiosyncratic, and not all patients or all pain syndromes respond to capsaicin (Immke and Gavva, 2006Immke D.C. Gavva N.R. Semin. Cell Dev. Biol. 2006; 17: 582-591Crossref PubMed Scopus (136) Google Scholar). The somewhat vague and diffuse traditional uses of this compound offer little assistance here, unlike artemisinin, for instance. Thus, testing for capsaicin efficacy is a matter of clinical trial and error, largely undermining the “tried and true” advantage of a traditional medicine. The major clinical advantage that capsaicin holds over other unrelated pain drugs under development is its approved status as a foodstuff. Like capsaicin, the polyphenol curcumin (Figure 1) is best known as a spice constituent: It is the yellow pigment component of the curry spice turmeric (Curcuma longa, known as haldi in Hindi). It is also, however, a drug used in Ayurveda and TCM in the treatment of diseases as diverse as rheumatism, fever, intestinal disorders, trauma, and amenorrhea (see the Analysis by S. Singh on page 765 of this issue). Modern research has attributed anti-inflammatory, immunomodulatory, antimalarial, and anticancer effects to this multitalented compound (Aggarwal et al., 2007Aggarwal B.B. Sundaram C. Malani N. Ichikawa H. Adv. Exp. Med. Biol. 2007; 595: 1-75Crossref PubMed Scopus (1128) Google Scholar). Like capsaicin, synthesis of curcumin is trivial and was first reported in 1910, but sufficient quantities of curcumin for therapeutic use are available from the spice. This is particularly important as low bioavailability of the parent compound coupled with rapid intestinal metabolism dictates large doses for clinical use (Sharma et al., 2005Sharma R.A. Gescher A.J. Steward W.P. Eur. J. Cancer. 2005; 41: 1955-1968Abstract Full Text Full Text PDF PubMed Scopus (1247) Google Scholar); derivatization of the natural product is actively being pursued. Given its pleiotropic clinical effects, it is perhaps not surprising that curcumin has documented effects on countless intracellular signaling pathways. Its anti-inflammatory action can be attributed largely to its inhibition of NF-κB activity, COX-2 and 5-LOX expression, and cytokine release (Aggarwal et al., 2007Aggarwal B.B. Sundaram C. Malani N. Ichikawa H. Adv. Exp. Med. Biol. 2007; 595: 1-75Crossref PubMed Scopus (1128) Google Scholar). Curcumin may directly target IκBα kinase to block NF-κB. It also binds to a number of other proteins, including thioredoxin reductase, several kinases, and several receptors (Aggarwal et al., 2007Aggarwal B.B. Sundaram C. Malani N. Ichikawa H. Adv. Exp. Med. Biol. 2007; 595: 1-75Crossref PubMed Scopus (1128) Google Scholar). The challenge here, then, as with many other natural products, is deciphering which of these targets is mechanistically valid for which biological activity. With such a broad spectrum of potential targets and activities described for curcumin, this is no easy task. Synthesis of derivatives that selectively ablate certain cellular and/or therapeutic effects is one possible route to tease apart this mechanism-function conundrum, perhaps in concert with radiolabeled fractionation experiments (as described above) or affinity chromatography with immobilized curcumin. The very versatility that makes curcumin appealing has also limited its rigorous clinical testing: There are wide-ranging efficacy reports, but most are based on preclinical, anecdotal, or pilot studies rather than on randomized, placebo-controlled, double-blind trials (Hsu and Cheng, 2007Hsu C.H. Cheng A.L. Adv. Exp. Med. Biol. 2007; 595: 471-480Crossref PubMed Scopus (308) Google Scholar). Activity has been reported in several inflammatory and autoimmune diseases and numerous cancers, both as a preventative agent and treatment, alone or in combination (Hsu and Cheng, 2007Hsu C.H. Cheng A.L. Adv. Exp. Med. Biol. 2007; 595: 471-480Crossref PubMed Scopus (308) Google Scholar). The relative ease and rapid payoff of undertaking preclinical or pilot studies, compared to rigorous clinical trials, has slowed the formal validation of curcumin. This is confounded by limited pharmaceutical company interest because curcumin itself is not patentable (although synthetic methods, derivatives, and pharmaceutical formulations are) and by the perception that, as a foodstuff, curcumin is more a nutraceutical (perhaps a dietary cancer preventative) than a traditional drug. This perception can only be changed by clinical studies showing successful disease treatment with curcumin. Phase I studies have documented tolerance up to 8000 mg/day, allowing a large dose-response range to be tested in phase II studies, several of which are underway for the treatment of cancer, psoriasis, and Alzheimer's disease (Table 1) (Hsu and Cheng, 2007Hsu C.H. Cheng A.L. Adv. Exp. Med. Biol. 2007; 595: 471-480Crossref PubMed Scopus (308) Google Scholar). We must await the outcomes of these studies before curcumin can be validated as a pharmaceutical. An effective drug should be facile and economical to produce and deliver, should display favorable absorption, distribution, metabolism, excretion, and toxicity (ADMET) characteristics, and should treat the targeted disease with specificity and efficacy. Traditional medicines, as with other natural products, can offer powerful leads for therapeutic development because (unlike synthetic libraries) they already have documented effects on the organism. However, the process from plant to product is a slow one. Despite the oft-shared limitations noted here, these five examples of traditional medicines are exceptional in the extent to which they have been studied and the success they have achieved in the clinic; countless other promising compounds wallow in obscurity. The challenges are formidable (Figure 1): Ethnopharmacologists must identify a medicine, its uses, and active components. These efforts are urgent, as traditional knowledge—and traditional plant species—are being lost at an alarming rate. Chemists must then synthesize the compound using a cost-effective method or develop alternative processes such as cell culture or transgenesis to enable useful-scale production. Despite continuing advances in synthetic chemistry, the very complexity of many natural products that is responsible for their desirable biological function can make production difficult. With a reliable supply of compound available, biologists can then identify and validate cellular targets and mechanisms of action. New tools are sorely needed for this particularly daunting challenge, such as methods that compare the phenotypic or gene expression profiles induced by a small molecule to those induced by known compounds (Hieronymus et al., 2006Hieronymus H. Lamb J. Ross K.N. Peng X.P. Clement C. Rodina A. Nieto M. Du J. Stegmaier K. Raj S.M. et al.Cancer Cell. 2006; 10: 321-330Abstract Full Text Full Text PDF PubMed Scopus (416) Google Scholar) or chemical enhancer/suppressor screens. Development of in silico tools to “dock” small molecules with protein structures to provide models for testing in vitro will likely come into their own with advances in structural genomics, as sufficient computational power becomes available. Ideally with a mechanism in hand, clinicians must then test the compound in the disease of interest (Table 1) while keeping an open mind for unexpected therapeutic activities and working with medicinal chemists to produce derivatives with improved ADMET properties. Finally, regulatory approval must be obtained, as with all drugs. This is particularly problematic if the active principle is an extract or mixture, rather than an isolated compound; the U.S. Food and Drug Administration has been understandably reluctant to approve multiple-agent drugs until recently (Schmidt et al., 2007Schmidt B.M. Ribnicky D.M. Lipsky P.E. Raskin I. Nat. Chem. Biol. 2007; 3: 360-366Crossref PubMed Scopus (243) Google Scholar). Only in 2006 was the first such drug approved: Polyphenon E (MediGene), a topical antiviral prepared from catechins extracted from green tea (Camellia sinensis). Artemisinin, triptolide, celastrol, capsaicin, and curcumin are “poster children” for the power and promise of turning traditional medicines into modern drugs. However, their stories highlight the ongoing interdisciplinary research efforts that continue to be necessary to realize the pharmaceutical potential of traditional therapeutics. C.M.C. is supported by the US National Institutes of Health (AI055194 and GM062120). T.W.C. is supported by a Canadian Institutes of Health Research Fellowship.
最长约 10秒,即可获得该文献文件

科研通智能强力驱动
Strongly Powered by AbleSci AI
更新
大幅提高文件上传限制,最高150M (2024-4-1)

科研通是完全免费的文献互助平台,具备全网最快的应助速度,最高的求助完成率。 对每一个文献求助,科研通都将尽心尽力,给求助人一个满意的交代。
实时播报
1秒前
红茸茸羊完成签到,获得积分10
1秒前
李爱国应助科研采纳,获得20
2秒前
3秒前
sptyzl发布了新的文献求助10
7秒前
wei发布了新的文献求助10
9秒前
10秒前
11秒前
12秒前
橙子发布了新的文献求助10
13秒前
starkisses完成签到,获得积分10
15秒前
兰西发布了新的文献求助10
16秒前
香蕉觅云应助于生有你采纳,获得10
16秒前
16秒前
所所应助请叫我过儿采纳,获得10
19秒前
情怀应助董可以采纳,获得10
20秒前
Fin2046发布了新的文献求助10
20秒前
22秒前
呆瓜发布了新的文献求助10
22秒前
冷傲世立发布了新的文献求助10
22秒前
wei完成签到,获得积分10
23秒前
23秒前
蓝冰发布了新的文献求助10
25秒前
29秒前
29秒前
于生有你发布了新的文献求助10
29秒前
30秒前
31秒前
31秒前
Ava应助兰西采纳,获得10
33秒前
养乐多发布了新的文献求助30
34秒前
34秒前
34秒前
34秒前
小黄包子发布了新的文献求助10
34秒前
milkcoffe发布了新的文献求助10
35秒前
35秒前
36秒前
董可以完成签到,获得积分20
37秒前
38秒前
高分求助中
请在求助之前详细阅读求助说明!!!! 20000
One Man Talking: Selected Essays of Shao Xunmei, 1929–1939 1000
Sphäroguß als Werkstoff für Behälter zur Beförderung, Zwischen- und Endlagerung radioaktiver Stoffe - Untersuchung zu alternativen Eignungsnachweisen: Zusammenfassender Abschlußbericht 1000
Yuwu Song, Biographical Dictionary of the People's Republic of China 700
[Lambert-Eaton syndrome without calcium channel autoantibodies] 520
The Three Stars Each: The Astrolabes and Related Texts 500
Additive Manufacturing Design and Applications 320
热门求助领域 (近24小时)
化学 材料科学 医学 生物 有机化学 工程类 生物化学 纳米技术 物理 内科学 计算机科学 化学工程 复合材料 遗传学 基因 物理化学 催化作用 电极 光电子学 量子力学
热门帖子
关注 科研通微信公众号,转发送积分 2467480
求助须知:如何正确求助?哪些是违规求助? 2135587
关于积分的说明 5441548
捐赠科研通 1860428
什么是DOI,文献DOI怎么找? 925290
版权声明 562645
科研通“疑难数据库(出版商)”最低求助积分说明 495006