Trastuzumab: Difference between revisions

From Wikipedia, the free encyclopedia
Content deleted Content added
m added "is"
Line 131: Line 131:
* [http://www.herceptin.com/herceptin/patient/index.jsp Herceptin] (manufacturer's website)
* [http://www.herceptin.com/herceptin/patient/index.jsp Herceptin] (manufacturer's website)
* [http://www.cancer.gov/cancertopics/druginfo/trastuzumab NCI Drug Information Summary for Patients]
* [http://www.cancer.gov/cancertopics/druginfo/trastuzumab NCI Drug Information Summary for Patients]
* [http://gentaur-worldwide.com/index.php?option=com_k2&view=item&id=78:trastuzumab-increases-the-relative-risk-of-brain-metastases&Itemid=530 Trastuzumab increases the relative risk of brain metastases]


{{Wiktionary}}
{{Wiktionary}}

Revision as of 11:19, 18 March 2013

Trastuzumab
Monoclonal antibody
TypeWhole antibody
SourceHumanized (from mouse)
TargetHER2/neu
Clinical data
Trade namesHerclon, Herceptin
AHFS/Drugs.comMonograph
Pregnancy
category
  • X
Routes of
administration
Intravenous
ATC code
Legal status
Legal status
Pharmacokinetic data
MetabolismUnknown, possibly reticuloendothelial system.
Elimination half-life2-12 days
Identifiers
CAS Number
DrugBank
ChemSpider
UNII
KEGG
ChEMBL
ECHA InfoCard100.224.377 Edit this at Wikidata
Chemical and physical data
FormulaC6470H10012N1726O2013S42
Molar mass145531.5 g/mol g·mol−1
 ☒NcheckY (what is this?)  (verify)

Trastuzumab (INN; trade names Herclon, Herceptin) is a monoclonal antibody that interferes with the HER2/neu receptor. Its main use is to treat certain breast cancers.

The HER receptors are proteins that are embedded in the cell membrane and communicate molecular signals from outside the cell (molecules called EGFs) to inside the cell, and turn genes on and off. The HER proteins stimulate cell proliferation. In some cancers, notably certain types of breast cancer, HER2 is over-expressed, and causes cancer cells to reproduce uncontrollably.[2]

The original studies of trastuzumab showed that it improved overall survival in late-stage (metastatic) breast cancer from 20.3 to 25.1 months.[2] In early stage breast cancer, it reduces the risk of cancer returning after surgery by an absolute risk of 9.5%, and the risk of death by an absolute risk of 3% however increases serious heart problems by an absolute risk of 2.1% which may resolve if treatment is stopped.[3] Trastuzumab is controversial partly because of its cost, as much as $54,000 per year,[4][5] and while certain private insurance companies in the U.S. and government health care systems in Canada, England and elsewhere have refused to pay for trastuzumab for certain patients, some companies have since accepted trastuzumab treatment as a covered preventative treatment.[6]

Trastuzumab is also being studied for the treatment of other cancers.[7] It has been used with some success in women with uterine papillary serous carcinomas that overexpress HER2/neu.[8]

Medical use

The original studies of trastuzumab showed that it improved overall survival in late-stage (metastatic) HER2-positive breast cancer from 20.3 to 25.1 months.[2] In early stage HER2-positive breast cancer, it reduces the risk of cancer returning after surgery by an absolute risk of 9.5%, and the risk of death by an absolute risk of 3% however increases serious heart problems by an absolute risk of 2.1% which may resolve if treatment is stopped.[3]

Trastuzumab has had a "major impact in the treatment of HER2-positive metastatic breast cancer".[9] The combination of trastuzumab with chemotherapy has been shown to increase both survival and response rate, in comparison to trastuzumab alone.[10]

It is possible to determine the "erbB2 status" of a tumor, which can be used to predict efficacy of treatment with trastuzumab. If it is determined that a tumor is overexpressing the erbB2 oncogene and the patient has no significant pre-existing heart disease, then a patient is eligible for treatment with trastuzumab.[11] It is surprising that although trastuzumab has great affinity for HER2 and high doses can be administered (due to its low toxicity), 70% of HER2+ patients do not respond to treatment. In fact resistance to the treatment develops rapidly, in virtually all patients. It is suggested that a mechanism of resistance is the lack of p27Kip1 translocation to the nucleus in some strains of cancer, enabling cdk2 to induce cell proliferation.[12]

Adverse effects

One of the significant complications of trastuzumab is its effect on the heart. Trastuzumab is associated with cardiac dysfunction in 2-7% of cases.[13] As a result, regular cardiac screening with either a MUGA scan or echocardiography is commonly undertaken during the trastuzumab treatment period.

Trastuzumab downregulates neuregulin-1 (NRG-1), which is essential for the activation of cell survival pathways in cardiomyocytes and the maintenance of cardiac function. NRG-1 activates the MAPK pathway and the PI3K/AKT pathway as well as focal adhesion kinases (FAK). These are all significant for the function and structure of cardiomyocytes. Trastuzumab can therefore lead to cardiac dysfunction.[14]

Approximately 10% of patients are unable to tolerate this drug because of pre-existing heart problems; physicians are balancing the risk of recurrent cancer against the higher risk of death due to cardiac disease in this population. The risk of cardiomyopathy is increased when trastuzumab is combined with anthracyclinechemotherapy (which itself is associated with cardiac toxicity).

If Herclon is used with a mixed dosage of Penicillin it can have side effects like numbness of the finger tips, cramps in the stomach. Also there might be a shock kind of sensation in the body. The body might get cold while the dosage is going on and the nerve through which it is being given would be warm. Your doctor would be informing you this prior to the dosage and would also get a MOU signed from you stating that you are aware about the effects of the mixed dosage.

Mechanism of action

The HER2 gene (also known as HER2/neu and ErbB2 gene) is amplified in 20-30% of early-stage breast cancers, which makes it overexpress epidermal growth factor (EGF) receptors in the cell membrane.[15] In some types of cancer HER2 may send signals without growth factors arriving and binding to the receptor, making its effect in the cell constitutive; however, trastuzumab is not effective in this case.

The HER2 pathway promotes cell growth and division when it is functioning normally; however when it is overexpressed, cell growth accelerates beyond its normal limits. In some types of cancer the pathway is exploited to promote rapid cell growth and proliferation and hence tumor formation.[16] The EGF pathway includes the receptors HER1 (EGFR), HER2, HER3, and HER4; the binding of EGF to HER is required to activate the pathway.[16] The pathway initiates the MAP Kinase pathway as well as the PI3 Kinase/AKT pathway, which in turn activates the NF-κB pathway.[17] In cancer cells the HER2 protein can be expressed up to 100 times more than in normal cells (2 million versus 20,000 per cell).[18] This overexpression leads to strong and constant proliferative signaling and hence tumor formation. Overexpression of HER2 also causes deactivation of checkpoints, allowing for even greater increases in proliferation.

HER2 extends across the cell membrane, and carries signals from outside the cell to the inside. Signaling compounds called mitogens (specifically EGF in this case) arrive at the cell membrane, and bind to the extracellular domain of the HER family of receptors. Those bound proteins then link (dimerize), activating the receptor. HER2 sends a signal from its intracellular domain, activating several different biochemical pathways. These include the PI3K/Akt pathway and the MAPK pathway. Signals on these pathways promote cell proliferation and the growth of blood vessels to nourish the tumor (angiogenesis).[19]

Normal cell division - mitosis - has checkpoints that keep cell division under control. Some of the proteins that control this cycle are called cdk2 (CDKs). Overexpression of HER2 sidesteps these checkpoints, causing cells to proliferate in an uncontrolled fashion.[12] This is caused by phosphorylation by Akt.

Trastuzumab binds to domain IV of the[20] extracellular segment of the HER2/neu receptor. Cells treated with trastuzumab undergo arrest during the G1 phase of the cell cycle so there is reduced proliferation. It has been suggested that trastuzumab induces some of its effect by downregulation of HER2/neu leading to disruption of receptor dimerization and signaling through the downstream PI3K cascade.[12] In addition, trastuzumab suppresses angiogenesis both by induction of antiangiogenic factors and repression of proangiogenic factors. It is thought that a contribution to the unregulated growth observed in cancer could be due to proteolytic cleavage of HER2/neu that results in the release of the extracellular domain. Trastuzumab has been shown to inhibit HER2/neu ectodomain cleavage in breast cancer cells.[21]

Experiments in laboratory animals indicate that antibodies, including trastuzumab, when bound to a cell, induce immune cells to kill that cell, and that such antibody-dependent cell-mediated cytotoxicity is another important mechanism of action.[22]

There may be other undiscovered mechanisms by which trastuzumab induces regression in cancer.

Predicting response to therapy

Trastuzumab inhibits the effects of overexpression of HER2. If the breast cancer doesn't overexpress HER2, trastuzumab will have no beneficial effect (and may cause harm). Doctors use laboratory tests to discover whether HER2 is overexpressed. In the routine clinical laboratory, the most commonly employed methods for this are immunohistochemistry (IHC) and either silver, chromogenic or fluorescent in situ hybridisation (SISH/CISH/FISH). HER2 amplification can be detected by virtual karyotyping of formalin-fixed paraffin embedded tumor. Virtual karyotyping has the added advantage of assessing copy number changes throughout the genome, in addition to detecting HER-2 amplification (but not overexpression). Numerous PCR-based methodologies have also been described in the literature.[23]

There are two FDA-approved commercial kits available for HER2 IHC; Dako HercepTest [24] and Ventana Pathway [1]. These are highly standardised, semi-quantitative assays which stratify expression levels into; 0 (<20,000 receptors per cell, no visible expression), 1+ (~100,000 receptors per cell, partial membrane staining, < 10% of cells overexpressing HER-2), 2+ (~500,000 receptors per cell, light to moderate complete membrane staining, > 10% of cells overexpressing HER-2), and 3+ (~2,000,000 receptors per cell, strong complete membrane staining, > 10% of cells overexpressing HER-2). The presence of cytoplasmic expression is disregarded. Treatment with trastuzumab is indicated in cases where HER2 expression has a score of 3+. However, IHC has been shown to have numerous limitations, both technical and interpretative, which have been found to impact on the reproducibility and accuracy of results, especially when compared with ISH methodologies. It is also true, however, that some reports have stated that IHC provides excellent correlation between gene copy number and protein expression.

Fluorescent in situ hybridization (FISH) is viewed as being the “gold standard” technique in identifying patients who would benefit from trastuzumab, but it is expensive and requires fluorescence microscopy and an image capture system. The main expense involved with CISH is in the purchase of FDA-approved kits, and as it is not a fluorescent technique it does not require specialist microscopy and slides may be kept permanently. Comparative studies of CISH and FISH have shown that these two techniques show excellent correlation. The lack of a separate chromosome 17 probe on the same section is an issue with regards to acceptance of CISH. As of June 2011 Roche has obtained FDA approval for the INFORM HER2 Dual ISH DNA Probe cocktail [25] developed by Ventana Medical Systems [2]. The DDISH (Dual-chromagen/Dual-hapten In-situ hybridization) cocktail uses both HER2 and Chromosome 17 hybridization probes for chromagenic visualization on the same tissue section. The detection can be achieved by using a combination of ultraView SISH(silver in-situ hybridization) and ultraView Red ISH for deposition of distinct chromgenic precipitates at the site of DNP or DIG labeled probes.[26]

Currently the recommended assays are a combination of IHC and FISH, whereby IHC scores of 0 and 1+ are negative (no trastuzumab treatment), scores of 3+ are positive (trastuzumab treatment), and score of 2+ (equivocal case) is referred to FISH for a definitive treatment decision. Industry best practices indicate the use of FDA-cleared Automated Tissue Image Systems by laboratories for automated processing of specimens, thereby reducing process variability, avoiding equivocal cases, and ensuring maximjum efficacy of trastuzumab therapy.

Optimal duration of adjuvant trastuzumab

The optimal duration of adjuvant trastuzumab is currently unknown. One year of treatment is generally accepted as the optimum length of therapy based on current clinical trial evidence that demonstrated the superiority of one year treatment over none.[27][28] However, a small Finnish trial also showed similar improvement with nine weeks of treatment over no therapy.[29] Due to the lack of direct head-to-head comparison in clinical trials, it is unknown whether a shorter duration of treatment may be just as effective (with fewer side effects) than the currently accepted practice of treatment for one year. Debate about treatment duration has become a relevant issue for many public health policy makers due to the high financial costs involved in the administration of trastuzumab for a year. Some countries with a taxpayer-funded public health system, such as New Zealand, have opted to fund only nine weeks of adjuvant therapy as a result.[30] Clinical trial data from Roche show that one year of therapy balances efficacy against adverse side effects.[31][32]

History

The biotech company Genentech developed trastuzumab jointly with UCLA and gained FDA approval in September 1998. The drug was first discovered by scientists including Dr. Axel Ullrich and Dr. H. Michael Shepard. At UCLA's Jonsson Cancer Center, Dr. Dennis Slamon subsequently worked on trastuzumab's development. A book about Dr. Slamon's work was made into a television film called Living Proof, that premiered in 2008.

Costs

Trastuzumab costs about US$70,000 for a full course of treatment,[33] Trastuzumab brought in $327 million in revenue for Genentech in the fourth quarter of 2007. Genentech refuses to explain the high cost of treatment.[34]

Australia has negotiated a lower price of A$50,000 per course of treatment.[35] Recently there has been controversy in New Zealand and the UK about public health funding of this drug in the adjuvant setting due to its high cost and perceived limited overall survival benefit (though not breast cancer-free survival benefit).[36] A case brought by a cancer victim in the UK succeeded on appeal after the local health authority refused to fund her treatment.[37][38] After a sustained campaign from cancer sufferers, the Ontario Ministry of Health in July 2005 decided that it would pay for treatments with trastuzumab and two other new and controversial anti-cancer drugs.[39]

Since October 2006 trastuzumab has been made available for Australian women and men with early stage breast cancer via the Pharmaceutical Benefits Scheme. This is estimated to cost the country over A$470 million for 4–5 years supply of the drug.[40]

Roche has inked a deal with Emcure in India to make an affordable version of this cancer drug available to the Indian market[41]

Roche has changed the trade name of the drug and has re-introduced an affordable version of the same in the Indian market. The new drug named Herclon would cost approximately RS 75,000 INR in the indian market. source?

See also

References

  1. ^ "FDA-sourced list of all drugs with black box warnings (Use Download Full Results and View Query links.)". nctr-crs.fda.gov. FDA. Retrieved 22 Oct 2023.
  2. ^ a b c Hudis, CA (2007). "Trastuzumab--mechanism of action and use in clinical practice". N Engl J Med. 357 (1): 39–51. doi:10.1056/NEJMra043186. PMID 17611206. Jul 5;357(1):39-51. Review /article
  3. ^ a b Moja L, Tagliabue L, Balduzzi S; et al. (2012). "Trastuzumab containing regimens for early breast cancer". Cochrane Database Syst Rev. 4: CD006243. doi:10.1002/14651858.CD006243.pub2. PMID 22513938. {{cite journal}}: Explicit use of et al. in: |author= (help)CS1 maint: multiple names: authors list (link) Cite error: The named reference "Cochrane2012" was defined multiple times with different content (see the help page).
  4. ^ "Cancer Studies Suggest One Year on Herceptin Is Best - WSJ.com".
  5. ^ "Current Length of Herceptin Treatment Is Supported by Studies - NYTimes.com".
  6. ^ "At last, Axa pays for Herceptin". 2006.
  7. ^ Vecchione L. Novel investigational drugs for gastric cancer. Expert Opin Investig Drugs. 2009 May 26. [Epub ahead of publication]. Review /article.
  8. ^ Santin AD, Bellone S, Roman JJ, McKenney JK, Pecorelli S. (2008). "Trastuzumab treatment in patients with advanced or recurrent endometrial carcinoma overexpressing HER2/neu". Int J Gynaecol Obstet. 102 (2): 128–31. doi:10.1016/j.ijgo.2008.04.008. PMID 18555254.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  9. ^ Tan, AR (2002). "Ongoing adjuvant trials with trastuzumab in breast cancer". Seminars in Oncology. 30 (5 Suppl 16): 54–64. doi:10.1053/j.seminoncol.2003.08.008. PMID 14613027. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  10. ^ Nahta, R (2003). "HER-2-Targeted Therapy –Lessons Learned and Future Directions". Clinical Cancer Research. 9 (14): 5078–5048. PMID 14613984. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  11. ^ Yu, D (2000). "Overexpression of ErbB2 in cancer and ErbB2-targeting strategies". Oncogene. 19 (53): 6115–6121. doi:10.1038/sj.onc.1203972. PMID 11156524. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  12. ^ a b c Kute, T (2004). "Development of Herceptin resistance in breast cancer cells". Cytometry. 57A (2): 86–93. doi:10.1002/cyto.a.10095. PMID 14750129. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  13. ^ Seidman, A; et al. (2002). "Cardiac Dysfunction in the Trastuzumab Clinical Trials Experience". Journal of Clinical Oncology. 20 (5): 1215–1221. doi:10.1200/JCO.20.5.1215. PMID 11870163. {{cite journal}}: Explicit use of et al. in: |author= (help)
  14. ^ Zeglinski, M., Ludke, A., Jassal, D. S. & Singal, P. K. Trastuzumab-induced cardiac dysfunction: A 'dual-hit'. Exp. Clin. Cardiol. 16, 70-74 (2011)
  15. ^ Bange, J (2001). "Molecular targets for breast cancer therapy and prevention". Nature Medicine. 7 (5): 548–552. doi:10.1038/87872. PMID 11329054. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  16. ^ a b "Targeted Therapies for Breast Cancer Tutorial". National Cancer Institute. Retrieved 19 April 2011.
  17. ^ Feldman, A M (28 March 2007). "Developing Strategies to Link Basic Cardiovascular Sciences with Clinical Drug Development: Another Opportunity for Translational Sciences". Clinical Pharmacology & Therapeutics. 81 (6): 887–892. doi:10.1038/sj.clpt.6100160. {{cite journal}}: |access-date= requires |url= (help); Unknown parameter |coauthors= ignored (|author= suggested) (help)
  18. ^ Winer, Eric. "HER2 Disease in the Metastatic and Adjuvant Settings". Medscape Education. Retrieved 20 April 2011.
  19. ^ Ménard, S (2003). "Biologic and therapeutic role of HER2 in cancer". Oncogene. 22 (42): 6570–6578. doi:10.1038/sj.onc.1206779. PMID 14528282. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  20. ^ http://www.nature.com/nature/journal/v421/n6924/full/nature01392.html
  21. ^ Albanell, J (2003). "Mechanism of action of anti-HER2 monoclonal antibodies: scientific update on trastuzumab and 2C4". Advances in Experimental Medicine and Biology. Advances in Experimental Medicine and Biology. 532: 253–268. doi:10.1007/978-1-4615-0081-0_21. ISBN 978-0-306-47762-1. PMID 12908564. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  22. ^ Clynes, RA; Towers, TL; Presta, LG; Ravetch, JV (2000). "Inhibitory Fc receptors modulate in vivo cytoxicity against tumor targets". Nat Med. 6 (4): 443–6. doi:10.1038/74704. PMID 10742152.
  23. ^ Jennings, B (1997). "A differential PCR assay for the detection of c-erbB 2 amplification used in a prospective study of breast cancer". Molecular Pathology. 50 (5): 254–256. doi:10.1136/mp.50.5.254. PMID 9497915. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  24. ^ http://www.dakousa.com/index/prod_search/prod_baseproducts.htm?productareaid=1&productgroupid=3&productsubgroupid=1003000
  25. ^ "Ventana Medical Systems, Inc. Receives FDA Approval for the First Fully Automated Diagnostic Assay for HER2 Gene Status Determination in Breast Cancer Patients". BioPortfolio.com. 2011-06-14. Retrieved 2013-01-06.
  26. ^ "Dual color dual hapten HER2 genotyping for breast biopsy specimens (DDISH): Concordance with fluorescence in situ hybridization (FISH)". ASCO. 2009-10-06. Retrieved 2013-01-06.
  27. ^ Romond, EH (2005). "Trastuzumab plus adjuvant chemotherapy for operable HER2-positive breast cancer". New England Journal of Medicine. 353 (16): 1673–1684. doi:10.1056/NEJMoa052122. PMID 16236738. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  28. ^ Piccart-Gebhart MJ, MJ (2005). "Trastuzumab after adjuvant chemotherapy in HER2-positive breast cancer". New England Journal of Medicine. 353 (16): 1659–1672. doi:10.1056/NEJMoa052306. PMID 16236737. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  29. ^ Joensuu H, Kellokumpu-Lehtinen PL, Bono P; et al. (2006). "Adjuvant docetaxel or vinorelbine with or without trastuzumab for breast cancer". N Engl J Med. 354 (8): 809–20. doi:10.1056/NEJMoa053028. PMID 16495393. {{cite journal}}: Explicit use of et al. in: |author= (help)CS1 maint: multiple names: authors list (link)
  30. ^ Metcalfe, S (2007). "PHARMAC funding of 9-week concurrent trastuzumab (Herceptin) for HER2-positive early breast cancer". N Z Med J. 120 (1256): 1U2593. PMID 17589560. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  31. ^ "Final analysis of Phase III HERA trial confirmed one year of Herceptin treatment as standard of care in early-stage HER2-positive breast cancer". Roche. Retrieved 2013-01-06.
  32. ^ "View - European Society for Medical Oncology". ESMO. Retrieved 2013-01-06.
  33. ^ Fleck L (2006). "The costs of caring: Who pays? Who profits? Who panders?". Hastings Cent Rep. 36 (3): 13–7. doi:10.1353/hcr.2006.0040. PMID 16776017.
  34. ^ "Will Herceptin Media Blitz Help Or Harm Patients?". Scoop. 2006-02-14. Retrieved 2006-12-01.
  35. ^ "Listing of Herceptin on PBS". Australian Government, Dept of Health and Ageing. 2006-10-01.
  36. ^ "Breast cancer drug to remain unfunded". New Zealand Herald. 2006-10-16. Retrieved 2006-12-01.
  37. ^ "The trastuzumab judgement". BBC News. 2006-04-12. Retrieved 2006-12-01.
  38. ^ "Update on Herceptin appraisal". National Institute for Health and Clinical Excellence. Retrieved 2006-12-01.
  39. ^ "Ontario to pay for cancer drugs". Canadian Broadcasting Corporation. 2005-07-22. Retrieved 2006-12-01.
  40. ^ Australian Government, Dept of Health and Ageing "Listing of Herceptin on PBS", 2006-10-1. http://www.health.gov.au/internet/main/publishing.nsf/Content/herceptin-govtdecision.htm
  41. ^ "Emcure signs deal to manufacture Roche's anti-cancer drugs". The Times Of India. 2012-03-02.

Further reading

External links