Camptothecin

Last updated
Camptothecin
Camptothecin.svg
Clinical data
ATC code
  • none
Identifiers
  • (S)-4-ethyl-4-hydroxy-1H-pyrano[3',4':6,7]indolizino[1,2-b]
    quinoline-3,14-(4H,12H)-dione
CAS Number
PubChem CID
DrugBank
ChemSpider
UNII
KEGG
ChEBI
ChEMBL
CompTox Dashboard (EPA)
ECHA InfoCard 100.113.172 OOjs UI icon edit-ltr-progressive.svg
Chemical and physical data
Formula C20H16N2O4
Molar mass 348.358 g·mol−1
3D model (JSmol)
Melting point 275 to 277 °C (527 to 531 °F)
  • O=C\1N4\C(=C/C2=C/1COC(=O)[C@]2(O)CC)c3nc5c(cc3C4)cccc5
  • InChI=1S/C20H16N2O4/c1-2-20(25)14-8-16-17-12(7-11-5-3-4-6-15(11)21-17)9-22(16)18(23)13(14)10-26-19(20)24/h3-8,25H,2,9-10H2,1H3/t20-/m0/s1 Yes check.svgY
  • Key:VSJKWCGYPAHWDS-FQEVSTJZSA-N Yes check.svgY
   (verify)

Camptothecin (CPT) is a topoisomerase inhibitor. It was discovered in 1966 by M. E. Wall and M. C. Wani in systematic screening of natural products for anticancer drugs. It was isolated from the bark and stem of Camptotheca acuminata (Camptotheca, Happy tree), a tree native to China used in traditional Chinese medicine. [1] [2] It has been used clinically more recently in China for the treatment of gastrointestinal tumors. [3] CPT showed anticancer activity in preliminary clinical trials, especially against breast, ovarian, colon, lung, and stomach cancers. [4] However, it has low solubility and adverse effects have been reported when used therapeutically, [3] so synthetic and medicinal chemists have developed numerous syntheses of camptothecin [5] [6] [7] and various derivatives to increase the benefits of the chemical, with good results. Four CPT analogues have been approved and are used in cancer chemotherapy [8] today: topotecan, irinotecan, belotecan, and trastuzumab deruxtecan. [9] [10] Camptothecin has also been found in other plants including Chonemorpha fragrans . [11]

Contents

Structures

CPT has a planar pentacyclic ring structure, that includes a pyrrolo[3,4-β]-quinoline moiety (rings A, B and C), conjugated pyridone moiety (ring D) and one chiral center at position 20 within the alpha- hydroxy lactone ring with (S) configuration (the E-ring). Its planar structure is thought to be one of the most important factors in topoisomerase inhibition. [12] [13]

Binding of CPT to topoisomerase I and DNA Camptothecin binding.svg
Binding of CPT to topoisomerase I and DNA

Binding

CPT binds to the topoisomerase I and DNA complex (the covalent complex) resulting in a ternary complex, and thereby stabilizing it. This prevents DNA re-ligation and therefore causes DNA damage which results in apoptosis. [14] CPT binds both to the enzyme and DNA with hydrogen bonds. The most important part of the structure is the E-ring which interacts from three different positions with the enzyme. The hydroxyl group in position 20 forms hydrogen bond to the side chain on aspartic acid number 533 (Asp533) in the enzyme. It is critical that the configuration of the chiral carbon is (S) because (R) is inactive. The lactone is bonded with two hydrogen bonds to the amino groups on arginine 364 (Arg364). The D-ring interacts with the +1 cytosine on non-cleaved strand and stabilizes the topoisomerase I-DNA covalent complex by forming hydrogen bond. This hydrogen bond is between carbonyl group in position 17 on the D-ring and amino group on the pyrimidine ring of +1 cytosine. [15] [16] CPT is selectively cytotoxic to the cells replicating DNA during S phase [17] and its toxicity is primarily a result of conversion of single-strand breaks into double-strand breaks when the replication fork collides with the cleavage complexes formed by DNA and CPT. [18]

Chemistry

The lactone ring in CPT is highly susceptible to hydrolysis. The open ring form is inactive and it must therefore be closed to inhibit topoisomerase I. The closed form is favored in acidic condition, as it is in many cancer cells microenvironment. CPT is transported into the cell by passive diffusion. Cellular uptake is favored by lipophilicity, which enhances intracellular accumulation. Lipophilicity makes compounds more stable because of improved lactone partitioning into red blood cells and consequently less hydrolysis of the lactone. CPT has affinity for human serum albumin (HSA), especially the carboxylate form of CPT. Because of that, the equilibrium between the lactone ring and the carboxylate form is driven toward the carboxylate. Reduced drug-HSA interactions [ disambiguation needed ] could result in improved activity. [15] [19]

Structure-activity relationship

Camptothecin Camptothecin numbering.svg
Camptothecin

Studies have shown that substitution at position 7, 9, 10 and 11 can have positive effect on CPT activity and physical properties, e.g. potency and metabolic stability. Enlargement of the lactone ring by one CH
2
unit
also enhances its abilities, as in homocamptothecin. Substitution at position 12 and 14 leads to inactive derivative. [19]

A- and B-ring modification

Alkyl substitution

Alkyl substitution at position 7 has shown increased cytotoxicity, such as ethyl (C2H5) or chloromethyl (CH2Cl). These groups are able to react with the DNA in the presence of topoisomerase I which leads to more tumor activity. It has also been shown that increasing the length of the carbon chain (in position 7) leads to increased lipophilicity and consequently greater potency and stability in human plasma. [15] [19] Other 7-modified CPT analogues are silatecans and karenitecins. They are potent inhibitors on topoisomerase I and both have alkylsilyl groups in position 7 which make them lipophilic and more stable. Silatecans or 7-silylcampthothecins have shown reduced drug-HSA interactions which contributes to its blood stability and they can also cross the blood brain barrier. DB-67 is a 10-hydroxy derivative and is among the most active silatecans. BNP1350 which belongs to the series of karenitecins exhibits cytotoxic activity and ability to overcome drug resistance. Still another route to make CPT’s lipophilic is to introduce lipophilic substituents, such as iminomethyl or oxyiminomethyl moieties. One of the most potent compounds is the oxyiminomethyl derivative ST1481 that has the advantage to overcome drug resistance caused by transport systems. [19] Basic nitrogen in a carbon chain at position 7 makes the compound more hydrophilic and hence more water-soluble. For example, is a derivate called CKD-602, which is a potent topoisomerase I inhibitor and successfully overcomes the poor water solubility and toxicity seen with CPT. [19] [20]

Considerably greater activity can be achieved by putting electron-withdrawing groups like amino, nitro, bromo or chloro at position 9 and 10 and hydroxyl group at position 10 or 11. But these compounds are relatively insoluble in aqueous solutions, which causes difficulty in administrations. Methoxy group at both position 10 and 11 simultaneously leads to inactivity. [12] [19]

Hexacyclic CPT analogues

Hexacyclic CPT analogues have shown great potency. For example, methylenedioxy or ethylenedioxy group connected between 10 and 11 form a 5 or 6 membered ring which leads to more water-soluble derivates and increased potency. Researches have shown that ethylenedioxy analogues are less potent than methylenedioxy. The reason is the unfavorable steric interactions of ethylenedioxy analogues with the enzyme. [12] [19]

Adding amino or chloro group at 9th position or chloromethyl group at 7th position to these 10, 11-methylenedioxy or ethylenedioxy analogues results in compounds with even greater cytotoxicity but weaker solubility in water. To yield 10, 11-methylenedioxy or ethylenedioxy analogues with good water solubility a good way is to introduce a water solubilising substituent at position 7. Lurtotecan meets those requirements; it’s a 10, 11-ethylenedioxy analogue with a 4-methylpiperazino-methylene at position 7 and has shown a great potency in clinical researches. [12]

A ring can also be formed between position 7 and 9, like position 10 and 11. That gives new opportunities to make water-soluble derivatives [5]. These hexacyclic CPT become more active when electron-withdrawing groups are put in position 11 and methyl or amino groups at 10. Exatecan is an example of hexacyclic CPT that has a 6 membered ring over position 7 and 9, and is 10-methyl, 11-fluoro substituted [4]. It is water-soluble and more potent than topotecan. [12] [19] [21]

C- and D-ring modification

The C- and D-rings have an essential role in the antitumor activity. Replacement in any position results in much less potent compound than parent compound in other cytotoxicity assay. [12]

Homocamptothecin Homocamptothecin.svg
Homocamptothecin

E-ring modifications

The E-ring doesn’t allow many structural changes without losing CPT activity because it is necessary for binding to the active site of TOP I. [22] One possible replacement is changing the hydroxyl group to Cl, F or Br because their polarizability is sufficient to stabilize the enzyme-complex. [19]

Another possible modification is to insert a methylene between hydroxyl and lactone on the E-ring yielding a seven membered β-hydroxylactone group, so-called homocamptothecin (hCPT). The hCPT’s hydroxyl has less inductive effect on the carboxyl group which makes the lactone very reactive. This enhances the interaction of the free hydroxyl group optimally with topoisomerase I and the covalent complex that forms in its presence are more stable. The E-ring of hCPT opens more slowly and the opening is irreversible. hCPTs exhibit enhanced human plasma stability because of decreased protein binding and more affinity for red blood cells than CPT. [12] [19]

CPT analogues

Since the discovery of CPT many analogues have been synthesized. Below is a schematic view of the CPT analogues that have been mentioned in the text above.

Camptothecin with radicals Camptothecin analogs.svg
Camptothecin with radicals
AnalogueR1R2R3R4
Topotecan —H Topotecan R2.svg —OH—H
Irinotecan (CPT-11) Irinotecan R1.svg —H Irinotecan R3.svg —H
Silatecan (DB-67, AR-67) Silatecan R1.svg —H—OH—H
Cositecan (BNP-1350) Cositecan R1.svg —H—H—H
Exatecan Exatecan R1R2.svg —CH3—F
Lurtotecan Lurtotecan R1.svg —H Lurtotecan R3R4.svg
Gimatecan (ST1481) Gimatecan R1.svg —H—H—H
Belotecan (CKD-602) Belotecan R1.svg —H—H—H
Rubitecan —H Rubitecan R2.svg —H—H

CPT is linked to a cyclodextrin-based polymer to form the investigational anti-cancer drug CRLX101. [23]

Biosynthesis

1 Tryptamine biosynthesis pathway 1 Tryptamine biosynthesis pathway.png
1 Tryptamine biosynthesis pathway
2 Secologanin biosynthesis pathway 2 Secologanin biosynthesis pathway.png
2 Secologanin biosynthesis pathway
Tryptamine and strictosidine to camptothecin Tryptamine and strictosidine to camptothecin.png
Tryptamine and strictosidine to camptothecin

Like all other monoterpenoid indole-alkaloids, biosynthesis of camptothecin requires production of the strictosidine. Strictosidine is synthesized through condensation reaction between tryptamine from shikimate pathway and secologanin from either mevalonate (MVA) pathway or non-mevalonate pathway (MEP). [24] Strictosidine then undergoes intermolecular cyclization to produce strictosamide, which is converted to camptothecin through a series of oxidation reactions by enzymes that still needs to be resolved. [25]

The shikimate pathway leading to biosynthesis of tryptamine is mostly understood. First, chorismate is converted to anthranilate by the alpha-subunit of anthranilate synthase (ASA). Anthranilate reacts with 5-phosphoribose pyrophosphate to produce 5-phosphoribosylanthranilate. Then this intermediate is converted to indole glycerol phosphate, which interacts with the alpha-subunit of tryptophan (TSA) synthase to yield indole. The beta-subunit of tryptophan synthase (TSB) catalyzes condensation of indole with serine, leading to tryptophan. In the next step, tryptamine is produced as the result of decarboxylation by tryptophan decarboxylase (TDC). [26]

Secologanin synthesis begins with condensation reaction between pyruvate and D-Glyceraldehyde-3-phosphate catalyzed by 1-deoxy-D-xylulose-5-phosphate synthase (DXS) to produce 1-deoxy-D-xylulose-5-phosphate (DXP). The conversion of DXP to isopentenyl diphosphate (IPP), which is the common terpenoid biosynthesis precursor involves 1-deoxy-D-xylulose-5-phosphate reductoisomerase (DXR) and 1-hydroxy-2-methyl-2(E)-butenyl-4-diphosphate reductase (HDR). The formation of IPP can be achieved by both MVA and MEP pathways. [26] Condensation of IPP and dimethylallyl diphosphate (DMAPP) yields geranyl diphosphate (GPP). The geraniol synthase (GS) then converts GPP to geraniol. [24] The conversion of geraniol to secologanin occurs through various enzymatic reactions. Based on studies with radioactive labelling and pathway specific inhibitors, MEP pathway is the primary source for secologanin. [26] Tryptamine from shikimate pathway and secologanin from MVA or MEP pathway are converted to strictosidine through a condensation reaction catalyzed by strictosidine synthase. Although it is not fully resolved, it has been postulated that camptothecin is produced from strictosidine via strictosamide, 3 (S)-pumiloside and 3 (S)-deoxypumiloside. [25]

Related Research Articles

<span class="mw-page-title-main">Epirubicin</span> Chemical compound

Epirubicin is an anthracycline drug used for chemotherapy. It can be used in combination with other medications to treat breast cancer in patients who have had surgery to remove the tumor. It is marketed by Pfizer under the trade name Ellence in the US and Pharmorubicin or Epirubicin Ebewe elsewhere.

<span class="mw-page-title-main">Irinotecan</span> Cancer medication

Irinotecan, sold under the brand name Camptosar among others, is a medication used to treat colon cancer, and small cell lung cancer. For colon cancer it is used either alone or with fluorouracil. For small cell lung cancer it is used with cisplatin. It is given intravenously.

<span class="mw-page-title-main">Topotecan</span> Chemical compound

Topotecan, sold under the brand name Hycamtin among others, is a chemotherapeutic agent medication that is a topoisomerase inhibitor. It is a synthetic, water-soluble analog of the natural chemical compound camptothecin. It is used in the form of its hydrochloride salt to treat ovarian cancer, lung cancer and other cancer types.

<span class="mw-page-title-main">Betulinic acid</span> Chemical compound

Betulinic acid is a naturally occurring pentacyclic triterpenoid which has antiretroviral, antimalarial, and anti-inflammatory properties, as well as a more recently discovered potential as an anticancer agent, by inhibition of topoisomerase. It is found in the bark of several species of plants, principally the white birch from which it gets its name, but also the ber tree, selfheal, the tropical carnivorous plants Triphyophyllum peltatum and Ancistrocladus heyneanus, Diospyros leucomelas, a member of the persimmon family, Tetracera boiviniana, the jambul, flowering quince, rosemary, and Pulsatilla chinensis.

Topoisomerase inhibitors are chemical compounds that block the action of topoisomerases, which are broken into two broad subtypes: type I topoisomerases (TopI) and type II topoisomerases (TopII). Topoisomerase plays important roles in cellular reproduction and DNA organization, as they mediate the cleavage of single and double stranded DNA to relax supercoils, untangle catenanes, and condense chromosomes in eukaryotic cells. Topoisomerase inhibitors influence these essential cellular processes. Some topoisomerase inhibitors prevent topoisomerases from performing DNA strand breaks while others, deemed topoisomerase poisons, associate with topoisomerase-DNA complexes and prevent the re-ligation step of the topoisomerase mechanism. These topoisomerase-DNA-inhibitor complexes are cytotoxic agents, as the un-repaired single- and double stranded DNA breaks they cause can lead to apoptosis and cell death. Because of this ability to induce apoptosis, topoisomerase inhibitors have gained interest as therapeutics against infectious and cancerous cells.

<span class="mw-page-title-main">Helenalin</span> Chemical compound

Helenalin, or (-)-4-Hydroxy-4a,8-dimethyl-3,3a,4a,7a,8,9,9a-octahydroazuleno[6,5-b]furan-2,5-dione, is a toxic sesquiterpene lactone which can be found in several plants such as Arnica montana and Arnica chamissonis Helenalin is responsible for the toxicity of the Arnica spp. Although toxic, helenalin possesses some in vitro anti-inflammatory and anti-neoplastic effects. Helenalin can inhibit certain enzymes, such as 5-lipoxygenase and leukotriene C4 synthase. For this reason the compound or its derivatives may have potential medical applications.

<span class="mw-page-title-main">TOP1</span> DNA topoisomerase enzyme

DNA topoisomerase 1 is an enzyme that in humans is encoded by the TOP1 gene. It is a DNA topoisomerase, an enzyme that catalyzes the transient breaking and rejoining of a single strand of DNA.

Strictosidine synthase (EC 4.3.3.2) is an enzyme in alkaloid biosynthesis that catalyses the condensation of tryptamine with secologanin to form strictosidine in a formal Pictet–Spengler reaction:

CRLX101 is an experimental approach to cancer chemotherapy that is under investigation in human trials. It is an example of a nanomedicine.

<span class="mw-page-title-main">Belotecan</span> Chemical compound

Belotecan is a drug used in chemotherapy. It is a semi-synthetic camptothecin analogue indicated for small-cell lung cancer and ovarian cancer, approved in South Korea under the trade name Camtobell, presented in 2 mg vials for injection. The drug has been marketed by ChongKunDang Pharmaceuticals since 2003.

<span class="mw-page-title-main">Ajmalicine</span> Chemical compound

Ajmalicine, also known as δ-yohimbine or raubasine, is an antihypertensive drug used in the treatment of high blood pressure. It has been marketed under numerous brand names including Card-Lamuran, Circolene, Cristanyl, Duxil, Duxor, Hydroxysarpon, Iskedyl, Isosarpan, Isquebral, Lamuran, Melanex, Raunatin, Saltucin Co, Salvalion, and Sarpan. It is an alkaloid found naturally in various plants such as Rauvolfia spp., Catharanthus roseus, and Mitragyna speciosa.

Tubulin inhibitors are chemotherapy drugs that interfere directly with the tubulin system, which is in contrast to those chemotherapy drugs acting on DNA. Microtubules play an important role in eukaryotic cells. Alpha- and beta-tubulin, the main components of microtubules, have gained considerable interest because of their function and biophysical properties and has become the subject of intense study. The addition of tubulin ligands can affect microtubule stability and function, including mitosis, cell motion and intracellular organelle transport. Tubulin binding molecules have generated significant interest after the introduction of the taxanes into clinical oncology and the general use of the vinca alkaloids. These compounds inhibit cell mitosis by binding to the protein tubulin in the mitotic spindle and preventing polymerization or depolymerization into the microtubules. This mode of action is also shared with another natural agent called colchicine.

The duocarmycins are members of a series of related natural products first isolated from Streptomyces bacteria in 1978. They are notable for their extreme cytotoxicity and thus represent a class of exceptionally potent antitumour antibiotics.

<span class="mw-page-title-main">Triacetic acid lactone</span> Chemical compound

Triacetic acid lactone is an organic compound derived enzymatically from glucose. It is a light yellow solid that is soluble in organic solvents.

<span class="mw-page-title-main">Strictosidine</span> Chemical compound

Strictosidine is a natural chemical compound and is classified as a glucoalkaloid and a vinca alkaloid. It is formed by the Pictet–Spengler condensation reaction of tryptamine with secologanin, catalyzed by the enzyme strictosidine synthase. Thousands of strictosidine derivatives are sometimes referred to by the broad phrase of monoterpene indole alkaloids. Strictosidine is an intermediate in the biosynthesis of numerous pharmaceutically valuable metabolites including quinine, camptothecin, ajmalicine, serpentine, vinblastine, vincristine and mitragynine.

Curacin A is a hybrid polyketide synthase (PKS)/nonribosomal peptide synthase (NRPS) derived natural product produced isolated from the cyanobacterium Lyngbya majuscula. Curacin A belongs to a family of natural products including jamaicamide, mupirocin, and pederin that have an unusual terminal alkene. Additionally, Curacin A contains a notable thiazoline ring and a unique cyclopropyl moiety, which is essential to the compound's biological activity. Curacin A has been characterized as potent antiproliferative cytotoxic compound with notable anticancer activity for several cancer lines including renal, colon, and breast cancer. Curacin A has been shown to interact with colchicine binding sites on tubulin, which inhibits microtubule polymerization, an essential process for cell division and proliferation.

<span class="mw-page-title-main">Discovery and development of gastrointestinal lipase inhibitors</span>

Lipase inhibitors belong to a drug class that is used as an antiobesity agent. Their mode of action is to inhibit gastric and pancreatic lipases, enzymes that play an important role in the digestion of dietary fat. Lipase inhibitors are classified in the ATC-classification system as A08AB . Numerous compounds have been either isolated from nature, semi-synthesized, or fully synthesized and then screened for their lipase inhibitory activity but the only lipase inhibitor on the market is orlistat . Lipase inhibitors have also shown anticancer activity, by inhibiting fatty acid synthase.

Fostriecin is a type I polyketide synthase (PKS) derived natural product, originally isolated from the soil bacterium Streptomyces pulveraceus. It belongs to a class of natural products which characteristically contain a phosphate ester, an α,β-unsaturated lactam and a conjugated linear diene or triene chain produced by Streptomyces. This class includes structurally related compounds cytostatin and phoslactomycin. Fostriecin is a known potent and selective inhibitor of protein serine/threonine phosphatases, as well as DNA topoisomerase II. Due to its activity against protein phosphatases PP2A and PP4 which play a vital role in cell growth, cell division, and signal transduction, fostriecin was looked into for its antitumor activity in vivo and showed in vitro activity against leukemia, lung cancer, breast cancer, and ovarian cancer. This activity is thought to be due to PP2A's assumed role in regulating apoptosis of cells by activating cytotoxic T-lymphocytes and natural killer cells involved in tumor surveillance, along with human immunodeficiency virus-1 (HIV-1) transcription and replication.

<span class="mw-page-title-main">Discovery and development of bisphosphonates</span> Drugs used to treat bone disorders

Bisphosphonates are an important class of drugs originally commercialised in the mid to late 20th century. They are used for the treatment of osteoporosis and other bone disorders that cause bone fragility and diseases where bone resorption is excessive. Osteoporosis is common in post-menopausal women and patients in corticosteroid treatment where biphosphonates have been proven a valuable treatment and also used successfully against Paget's disease, myeloma, bone metastases and hypercalcemia. Bisphosphonates reduce breakdown of bones by inhibiting osteoclasts, they have a long history of use and today there are a few different types of bisphosphonate drugs on the market around the world.

<span class="mw-page-title-main">Diflomotecan</span> Chemical compound

Diflomotecan is a chemotherapeutic agent that is a topoisomerase inhibitor (Top1). It varies from other camptothecin based Top1 inhibitors like topotecan in having a 7-membered E-ring. The oxepan-2-one ring in the homocamptothecin analogues is more stable in plasma compared to the 6-membered lactone in camptothecin. Diflomotecan was the first homocamptothecin to enter clinical trials and is currently in phase I for treating patients with sensitive small cell lung cancer (SCLC).

References

  1. Govindachari TR, Viswnathan N (1972). "the stem bark of Mappia foetida, a tree native to India, has proved to be another source significant for the isolation of camptothecin". Phytochemistry. 11 (12): 3529–31. doi:10.1016/s0031-9422(00)89852-0.
  2. Efferth T, Fu YJ, Zu YG, Schwarz G, Konkimalla VS, Wink M (2007). "Molecular target-guided tumor therapy with natural products derived from traditional Chinese medicine". Current Medicinal Chemistry. 14 (19): 2024–2032. doi:10.2174/092986707781368441. PMID   17691944.
  3. 1 2 "Camptothecin". Chemnetbase - Dictionary of Drugs.
  4. Wang XH, Huang M, Zhao CK, Li C, Xu L (June 2019). "Design, synthesis, and biological activity evaluation of campthothecin-HAA-Norcantharidin conjugates as antitumor agents in vitro". Chemical Biology & Drug Design. 93 (6): 986–992. doi:10.1111/cbdd.13397. PMID   30218487. S2CID   52277958.
  5. Reich HJ. "Curran Synthesis of Camptothecin". Archived from the original on 2009-09-05.
  6. Reich HJ. "Comins Synthesis of Camptothecin". Archived from the original on 2009-09-05.
  7. Reich HJ. "Rapaport Synthesis of Camptothecin". Archived from the original on September 7, 2009.
  8. Takimoto CH, Calvo E (2008). "Principles of Oncologic Pharmacotherapy". In Pazdur R, Wagman LD, Camphausen KA, Hoskins WJ (eds.). Cancer Management: A Multidisciplinary Approach (11th ed.). Archived from the original on 15 May 2009.
  9. Wall ME, Wani MC, Cook CA, Palmer KH, McPhail AA, Sim GA (1966). "Plant antitumor agents. I. The isolation and structure of camptothecin, a novel alkaloidal leukemia and tumor inhibitor from camptotheca acuminate". Journal of the American Chemical Society . 88 (16): 3888–3890. doi:10.1021/ja00968a057.
  10. Samuelsson G (2004). Drugs of Natural Origin: a Textbook of Pharmacognosy (5 ed.). Stokkholm: Swedish pharmaceutical press. ISBN   91-974318-4-2.
  11. Isah T, Umar S (September 2018). "Influencing in vitro clonal propagation of Chonemorpha fragrans (moon) Alston by culture media strength, plant growth regulators, carbon source and photo periodic incubation". Journal of Forestry Research. 31: 27–43. doi:10.1007/s11676-018-0794-3. S2CID   52297102.
  12. 1 2 3 4 5 6 7 Ulukan H, Swaan PW (2002). "Camptothecins: a review of their chemotherapeutic potential". Drugs (27 ed.). 62 (14): 2039–2057. doi:10.2165/00003495-200262140-00004. PMID   12269849.
  13. Lu AJ, Zhang ZS, Zheng MY, Zou HJ, Luo XM, Jiang HL (February 2007). "3D-QSAR study of 20 (S)-camptothecin analogs". Acta Pharmacologica Sinica. 28 (2): 307–314. doi: 10.1111/j.1745-7254.2007.00477.x . PMID   17241535.
  14. "Camptothecin". DrugBank. Retrieved 9 October 2016.
  15. 1 2 3 Adams DJ, Wahl ML, Flowers JL, Sen B, Colvin M, Dewhirst MW, et al. (January 2006). "Camptothecin analogs with enhanced activity against human breast cancer cells. II. Impact of the tumor pH gradient". Cancer Chemotherapy and Pharmacology. 57 (2): 145–154. doi:10.1007/s00280-005-0008-5. PMID   16001167. S2CID   23652115.
  16. Redinbo MR, Stewart L, Kuhn P, Champoux JJ, Hol WG (March 1998). "Crystal structures of human topoisomerase I in covalent and noncovalent complexes with DNA". Science. 279 (5356): 1504–1513. Bibcode:1998Sci...279.1504R. doi:10.1126/science.279.5356.1504. PMID   9488644.
  17. Del Bino G, Lassota P, Darzynkiewicz Z (March 1991). "The S-phase cytotoxicity of camptothecin". Experimental Cell Research. 193 (1): 27–35. doi:10.1016/0014-4827(91)90534-2. PMID   1995300.
  18. Pommier Y, Redon C, Rao VA, Seiler JA, Sordet O, Takemura H, et al. (November 2003). "Repair of and checkpoint response to topoisomerase I-mediated DNA damage". Mutation Research. 532 (1–2): 173–203. doi:10.1016/j.mrfmmm.2003.08.016. PMID   14643436.
  19. 1 2 3 4 5 6 7 8 9 10 Zunino F, Dallavalleb S, Laccabuea D, Berettaa G, Merlinib L, Pratesi G (2002). "Current status and perspectives in the development of camptothecins". Current Pharmaceutical Design (27 ed.). 8 (27): 2505–2520. doi:10.2174/1381612023392801. PMID   12369944.
  20. Chung MK, Han SS, Kim JC (August 2006). "Evaluation of the toxic potentials of a new camptothecin anticancer agent CKD-602 on fertility and early embryonic development in rats". Regulatory Toxicology and Pharmacology. 45 (3): 273–281. doi:10.1016/j.yrtph.2006.05.004. PMID   16814440.
  21. Palumbo M, Sissi C, Gatto B, Moro S, Zagotto G (November 2001). "Quantitation of camptothecin and related compounds". Journal of Chromatography. B, Biomedical Sciences and Applications. 764 (1–2): 121–140. doi:10.1016/S0378-4347(01)00345-0. PMID   11817024.
  22. Venditto VJ, Simanek EE (April 2010). "Cancer therapies utilizing the camptothecins: a review of the in vivo literature". Molecular Pharmaceutics. 7 (2): 307–349. doi:10.1021/mp900243b. PMC   3733266 . PMID   20108971.
  23. "Cerulean Raises $24M to Progress Clinical Development of Nanopharmaceuticals". 15 Nov 2010.
  24. 1 2 Yamazaki Y, Kitajima M, Arita M, Takayama H, Sudo H, Yamazaki M, et al. (January 2004). "Biosynthesis of camptothecin. In silico and in vivo tracer study from [1-13C]glucose". Plant Physiology. 134 (1): 161–170. doi: 10.1104/pp.103.029389 . PMC   316296 . PMID   14657405.
  25. 1 2 Lorence A, Nessler CL (October 2004). "Camptothecin, over four decades of surprising findings". Phytochemistry. 65 (20): 2735–2749. Bibcode:2004PChem..65.2735L. doi:10.1016/j.phytochem.2004.09.001. PMID   15474560.
  26. 1 2 3 New Light on Alkaloid Biosynthesis and Future Prospects. Academic Press. 27 July 2013. pp. 143–149. ISBN   978-0-08-099411-6.