1
|
Smith AF, Klotz A, Wormstone IM. Improving the drug development process by reducing the impact of adverse events: the case of cataracts considered. Drug Discov Today 2016; 21:510-6. [PMID: 26775751 DOI: 10.1016/j.drudis.2016.01.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Revised: 12/03/2015] [Accepted: 01/04/2016] [Indexed: 12/11/2022]
Abstract
Cataract was used as a model for the prevalence and economic impact of adverse events during the drug development process. Meta-analysis revealed a reported prevalence of cataract at 12.0% (1.0-43.3%), 3.8% (2.4-12.5%), 1.0% (0.0-8.1%), 1.7% (0.0-34.8%) and 3.8% (2.3-5.7%) of compounds in preclinical, Phase I, II, III and IV clinical trials, respectively. Utilising a human-based in vitro screening assay to predict cataractogenic potential in human could allow better selection of novel compounds at early-stage drug development. This could significantly reduce costs and ultimately increase the probability of a drug obtaining FDA approval for a clinical application.
Collapse
Affiliation(s)
- Andrew F Smith
- MedMetrics Inc., Ottawa, Canada; Department of Ophthalmology, King's College London, London, UK.
| | | | | |
Collapse
|
2
|
Nikolov IG, Joki N, Galmiche A, Nguyen-Khoa T, Guerrera IC, Guillonneau F, Ivanovski O, Phan O, Maizel J, Marçon F, Benchitrit J, Lucas A, Edelman A, Lacour B, Drüeke TB, Massy ZA. Farnesyltransferase inhibitor R115777 protects against vascular disease in uremic mice. Atherosclerosis 2013; 229:42-51. [DOI: 10.1016/j.atherosclerosis.2013.02.041] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2012] [Revised: 02/08/2013] [Accepted: 02/25/2013] [Indexed: 11/26/2022]
|
3
|
A phase II trial of capecitabine in combination with the farnesyltransferase inhibitor tipifarnib in patients with anthracycline-treated and taxane-resistant metastatic breast cancer: an Eastern Cooperative Oncology Group Study (E1103). Breast Cancer Res Treat 2012; 134:345-52. [PMID: 22547107 DOI: 10.1007/s10549-012-2071-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2012] [Accepted: 04/16/2012] [Indexed: 10/28/2022]
Abstract
Capecitabine produces an objective response rate of up to 25% in anthracycline-treated, taxane-resistant metastatic breast cancer (MBC). The farnesyltransferase inhibitor tipifarnib inhibits Ras signaling and has clinical activity when used alone in MBC. The objective of this study was to determine the efficacy and safety of tipifarnib-capecitabine combination in MBC patients who were previously treated with an anthracycline and progressed on taxane therapy. Eligible patients received oral capecitabine 1,000 mg/m2 twice daily plus oral tipifarnib 300 mg twice daily on days 1-14 every 21 days. The primary endpoint was ORR. The trial was powered to detect an improvement in response rate from 25 to 40%. Among 63 eligible, partial response occurred in six patients (9.5%; 90% CI 4.2-17.9%), median progression-free survival was 2.6 months (95% CI 2.1-4.4), and median overall survival was 11.4 months (95% CI 7.7-14.0). Dose modifications were required for 43 patients (68%) for either tipifarnib and/or capecitabine. Grades 3 and 4 toxicities were seen in 30 patients (44%; 90% CI 44.4-67.0%) and 11 patients (16%; 90% CI 10.8-29.0%), respectively. The most common grade 3 toxicities included neutropenia, nausea, and vomiting; and the most common grade 4 toxicity was neutropenia (8 out of 11 cases). The tipifarnib-capecitabine combination is not more effective than capecitabine alone in MBC patients who were previously treated with an anthracycline and taxane therapy.
Collapse
|
4
|
Wells EM, Rao AAN, Scafidi J, Packer RJ. Neurotoxicity of biologically targeted agents in pediatric cancer trials. Pediatr Neurol 2012; 46:212-21. [PMID: 22490765 PMCID: PMC3626408 DOI: 10.1016/j.pediatrneurol.2012.02.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2011] [Accepted: 02/10/2012] [Indexed: 02/07/2023]
Abstract
Biologically targeted agents offer the promise of delivering specific anticancer effects while limiting damage to healthy tissue, including the central and peripheral nervous systems. During the past 5-10 years, these agents were examined in preclinical and adult clinical trials, and are used with increasing frequency in children with cancer. This review evaluates current knowledge about neurotoxicity from biologically targeted anticancer agents, particularly those in pediatric clinical trials. For each drug, neurotoxicity data are reviewed in adult (particularly studies of brain tumors) and pediatric studies when available. Overall, these agents are well tolerated, with few serious neurotoxic effects. Data from younger patients are limited, and more neurotoxicity may occur in the pediatric population because these agents target pathways that control not only tumorigenesis but also neural maturation. Further investigation is needed into long-term neurologic effects, particularly in children.
Collapse
Affiliation(s)
- Elizabeth M. Wells
- Brain Tumor Institute, Children's National Medical Center, Washington, DC
- Center for Neuroscience and Behavioral Medicine, Children's National Medical Center, Washington, DC
- Department of Neurology and Pediatrics, George Washington University, Washington, DC
| | - Amulya A. Nageswara Rao
- Brain Tumor Institute, Children's National Medical Center, Washington, DC
- Center for Neuroscience and Behavioral Medicine, Children's National Medical Center, Washington, DC
- Department of Neurology and Pediatrics, George Washington University, Washington, DC
- Division of Pediatric Hematology/Oncology, Department of Pediatrics and Adolescent Medicine, Mayo Clinic, Rochester, Minnesota
| | - Joseph Scafidi
- Brain Tumor Institute, Children's National Medical Center, Washington, DC
- Center for Neuroscience and Behavioral Medicine, Children's National Medical Center, Washington, DC
- Department of Neurology and Pediatrics, George Washington University, Washington, DC
| | - Roger J. Packer
- Brain Tumor Institute, Children's National Medical Center, Washington, DC
- Center for Neuroscience and Behavioral Medicine, Children's National Medical Center, Washington, DC
- Department of Neurology and Pediatrics, George Washington University, Washington, DC
- Communications should be addressed to: Dr. Packer; Department of Neurology; Children's National Medical Center; 111 Michigan Avenue NW; Washington, DC 20010.
| |
Collapse
|
5
|
Wong NS, Meadows KL, Rosen LS, Adjei AA, Kaufmann SH, Morse MA, Petros WP, Zhu Y, Statkevich P, Cutler DL, Meyers ML, Hurwitz HI. A phase I multicenter study of continuous oral administration of lonafarnib (SCH 66336) and intravenous gemcitabine in patients with advanced cancer. Cancer Invest 2012; 29:617-25. [PMID: 22011284 DOI: 10.3109/07357907.2011.621912] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
We conducted a phase I study to assess safety, pharmacokinetics, pharmacodynamics, and activity of lonafarnib plus gemcitabine. Subjects received oral lonafarnib twice daily and gemcitabine on days 1, 8, and 15 every 28 days; multiple dose levels were explored. Lonafarnib had no apparent effect on gemcitabine PK. Mean lonafarnib half-life ranged from 4 to 7 hr; median T(max) values ranged from 4 to 8 hr. Two patients had partial response; seven patients had stable disease at least 6 months. Oral lonafarnib at 150 mg a.m./100 mg p.m. plus gemcitabine at 1,000 mg/m(2) is the maximum tolerated dose with acceptable safety and tolerability.
Collapse
Affiliation(s)
- Nan Soon Wong
- Duke University Medical Center, Durham, North Carolina, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
6
|
Nghiemphu PL, Wen PY, Lamborn KR, Drappatz J, Robins HI, Fink K, Malkin MG, Lieberman FS, DeAngelis LM, Torres-Trejo A, Chang SM, Abrey L, Fine HA, Demopoulos A, Lassman AB, Kesari S, Mehta MP, Prados MD, Cloughesy TF. A phase I trial of tipifarnib with radiation therapy, with and without temozolomide, for patients with newly diagnosed glioblastoma. Int J Radiat Oncol Biol Phys 2010; 81:1422-7. [PMID: 20934264 DOI: 10.1016/j.ijrobp.2010.07.1997] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2010] [Revised: 07/14/2010] [Accepted: 07/16/2010] [Indexed: 10/19/2022]
Abstract
PURPOSE To determine the maximum tolerated dose (MTD) of tipifarnib in combination with conventional radiotherapy for patients with newly diagnosed glioblastoma. The MTD was evaluated in three patient cohorts, stratified based on concurrent use of enzyme-inducing antiepileptic drugs (EIAED) or concurrent treatment with temozolomide (TMZ): Group A: patients not receiving EIAED and not receiving TMZ; Group A-TMZ: patients not receiving EIAED and receiving treatment with TMZ; Group B: any patients receiving EIAED but not TMZ. PATIENTS AND METHODS After diagnostic surgery or biopsy, treatment with tipifarnib started 5 to 9 days before initiating radiotherapy, twice daily, in 4-week cycles using discontinuous dosing (21 out of 28 days), until toxicity or progression. For Group A-TMZ, patients also received TMZ daily during radiotherapy and then standard 5/28 days dosing after radiotherapy. Dose-limiting toxicity (DLT) was determined over the first 10 weeks of therapy for all cohorts. RESULTS Fifty-one patients were enrolled for MTD determination: 10 patients in Group A, 21 patients in Group A-TMZ, and 20 patients in Group B. In the Group A and Group A-TMZ cohorts, patients achieved the intended MTD of 300 mg twice daily (bid) with DLTs including rash and fatigue. For Group B, the MTD was determined as 300 mg bid, half the expected dose. The DLTs included rash and one intracranial hemorrhage. Thirteen of the 20 patients evaluated in Group A-TMZ were alive at 1 year. CONCLUSION Tipifarnib is well tolerated at 300 mg bid given discontinuously (21/28 days) in 4-week cycles, concurrently with standard chemo/radiotherapy. A Phase II study should evaluate the efficacy of tipifarnib with radiation and TMZ in patients with newly diagnosed glioblastoma and not receiving EIAED.
Collapse
|
7
|
Mitra A, Shevde LA, Samant RS. Multi-faceted role of HSP40 in cancer. Clin Exp Metastasis 2009; 26:559-67. [PMID: 19340594 DOI: 10.1007/s10585-009-9255-x] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2008] [Accepted: 03/12/2009] [Indexed: 12/25/2022]
Abstract
HSP40 (DNAJ) is an understudied family of co-chaperones. The human genome codes for over 41 members of HSP40 family that reside at distinct intracellular locations. Despite their large numbers, little is known about their physiologic roles. Recent research has revealed involvement of some of the DNAJ family members in various types of cancers. In this article we summarize the information about the involvement of human DNAJ family members in various aspects of cancer biology. Furthermore we discuss the potential role of the J domain of DNAJ proteins in cancer biology.
Collapse
Affiliation(s)
- Aparna Mitra
- Department of Oncologic Sciences, Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36604, USA
| | | | | |
Collapse
|
8
|
Chow LQM, Eckhardt SG, O’Bryant CL, Schultz MK, Morrow M, Grolnic S, Basche M, Gore L. A phase I safety, pharmacological, and biological study of the farnesyl protein transferase inhibitor, lonafarnib (SCH 663366), in combination with cisplatin and gemcitabine in patients with advanced solid tumors. Cancer Chemother Pharmacol 2008; 62:631-46. [PMID: 18058098 PMCID: PMC2813768 DOI: 10.1007/s00280-007-0646-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2007] [Accepted: 11/12/2007] [Indexed: 10/22/2022]
Abstract
PURPOSE This phase I study was conducted to evaluate the safety, tolerability, pharmacological properties and biological activity of the combination of the lonafarnib, a farnesylproteintransferase (FTPase) inhibitor, with gemcitabine and cisplatin in patients with advanced solid malignancies. EXPERIMENTAL DESIGN This was a single institution study to determine the maximal tolerated dose (MTD) of escalating lonafarnib (75-125 mg po BID) with gemcitabine (750-1,000 mg/m(2) on days 1, 8, 15) and fixed cisplatin (75 mg/m(2) day 1) every 28 days. Due to dose-limiting toxicities (DLTs) of neutropenia and thrombocytopenia in initial patients, these patients were considered "heavily pre-treated" and the protocol was amended to limit prior therapy and re-escalate lonafarnib in "less heavily pre-treated patients" on 28-day and 21-day schedules. Cycle 1 and 2 pharmacokinetics (PK), and farnesylation of the HDJ2 chaperone protein and FPTase activity were analyzed. RESULTS Twenty-two patients received 53 courses of therapy. Nausea, vomiting, and fatigue were frequent in all patients. Severe toxicities were observed in 91% of patients: neutropenia (41%), nausea (36%), thrombocytopenia (32%), anemia (23%) and vomiting (23%). Nine patients withdrew from the study due to toxicity. DLTs of neutropenia, febrile neutropenia, thrombocytopenia, and fatigue limited dose-escalation on the 28-day schedule. The MTD was established as lonafarnib 75 mg BID, gemcitabine 750 mg/m(2) days 1, 8, 15, and cisplatin 75 mg/m(2) in heavily pre-treated patients. The MTD in the less heavily pre-treated patients could not be established on the 28-day schedule as DLTs were observed at the lowest dose level, and dose escalation was not completed on the 21-day schedule due to early study termination by the Sponsor. No PK interactions were observed. FTPase inhibition was not observed at the MTD, however HDJ-2 gel shift was observed in one patient at the 100 mg BID lonafarnib dose. Anti-cancer activity was observed: four patients had stable disease lasting >2 cycles, one subject had a complete response, and another had a partial response, both with metastatic breast cancer. CONCLUSION Lonafarnib 75 mg BID, gemcitabine 750 mg/m(2) days 1, 8, 15, and cisplatin 75 mg/m(2) day 1 on a 28-day schedule was established as the MTD. Lonafarnib did not demonstrate FTPase inhibition at these doses. Despite the observed efficacy, substantial toxicity and questionable contribution of anti-tumor activity of lonafarnib to gemcitabine and cisplatin limits further exploration of this combination.
Collapse
Affiliation(s)
| | | | | | | | - Mark Morrow
- University of Colorado Cancer Center, Aurora, CO, USA
| | - Stacy Grolnic
- University of Colorado Cancer Center, Aurora, CO, USA
| | | | - Lia Gore
- University of Colorado Cancer Center, Aurora, CO, USA
- The Children’s Hospital, Denver, CO, USA
- University of Colorado Health Sciences Center at Fitzsimons, Mail Stop 8302, PO Box 6511, Aurora, CO 80045, USA
| |
Collapse
|
9
|
Besnard T, Renée N, Etienne-Grimaldi MC, François E, Milano G. Optimized blood sampling with cytidine deaminase inhibitor for improved analysis of capecitabine metabolites. J Chromatogr B Analyt Technol Biomed Life Sci 2008; 870:117-20. [PMID: 18562256 DOI: 10.1016/j.jchromb.2008.05.040] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2007] [Revised: 05/02/2008] [Accepted: 05/08/2008] [Indexed: 11/15/2022]
Abstract
The 5FU prodrug capecitabine undergoes a 3-step enzymatic conversion, including the conversion of 5'DFRC into 5'DFUR by cytidine deaminase (CDA). The presence of CDA activity in blood led us to analyze the possible ex vivo conversion of 5'DFCR into 5'DFUR in blood samples. We thus examined the impact of the addition of a CDA inhibitor (tetrahydrouridine (THU) 1 microM final) in blood. Blood samples from 3 healthy volunteers were taken on tubes containing or not THU. Blood was spiked with 5'DFCR (20 microM final) (T0) and was maintained at room temperature for 2 h. Plasma concentrations of 5'DFRC and 5'DFUR were analyzed with an optimized HPLC assay. In the absence of THU, 5'DFUR was detectable as early as T0. The percent of 5'DFUR produced relative to 5'DFCR increased over time, up to 7.7 % at 2h. In contrast, the presence of THU totally prevents the formation of 5'DFUR. The impact of THU for preventing the conversion of 5'DFCR was confirmed by the analysis of blood samples from 2 capecitabine-treated patients. Addition of THU in the sampling-tube before the introduction of blood is thus strongly recommended in order to guarantee accurate conditions for reliable measurement of capecitabine metabolites in plasma, and thus faithful pharmacokinetic data.
Collapse
|
10
|
Martin LA, Head JE, Pancholi S, Salter J, Quinn E, Detre S, Kaye S, Howes A, Dowsett M, Johnston SRD. The farnesyltransferase inhibitor R115777 (tipifarnib) in combination with tamoxifen acts synergistically to inhibit MCF-7 breast cancer cell proliferation and cell cycle progression in vitro and in vivo. Mol Cancer Ther 2007; 6:2458-67. [PMID: 17876043 DOI: 10.1158/1535-7163.mct-06-0452] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Cross-talk between receptor tyrosine kinases and estrogen receptor is at least partly responsible for the development of acquired resistance to endocrine therapies. Hence, targeting receptor tyrosine kinases and their downstream partners with inhibitors/antagonists may reverse this resistance. Although ras mutations are rare in breast cancer (2%), aberrant function of Ras signal transduction pathways is common. We therefore investigated the efficacy of the farnesyltransferase inhibitor (FTI) R115777 (tipifarnib) in combination with tamoxifen in MCF-7 human breast cancer models both in vitro and in vivo. There was a synergistic antiproliferative interaction between R115777 and 4-hydroxy-tamoxifen in vitro as calculated by median effect analysis. The combination resulted in a significantly greater G(1) arrest than either drug alone and this was associated with marked inhibition of cyclin D1 and induction of the cell cycle inhibitor p27(kip1). Combining R115777 with either tamoxifen or estrogen withdrawal in vivo produced a significantly greater inhibition of tumor growth and lower xenograft cell proliferation than either therapy alone. These results suggest that the combination of this FTI with endocrine therapy may be of therapeutic benefit in the treatment of breast cancer. Enhanced G1 arrest due to modulation of cell cycle regulatory proteins may be the underlying mechanism for the positive interaction between FTIs and tamoxifen.
Collapse
Affiliation(s)
- Lesley-Ann Martin
- Breakthrough Breast Cancer Centre, Institute of Cancer Research, Fulham Road, London, SW3 6JB United Kingdom.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Findlay M, von Minckwitz G, Wardley A. Effective oral chemotherapy for breast cancer: pillars of strength. Ann Oncol 2007; 19:212-22. [PMID: 18006898 DOI: 10.1093/annonc/mdm285] [Citation(s) in RCA: 94] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Traditionally, anticancer therapy has been dominated by intravenous drug therapy. However, oral agents provide an attractive approach to chemotherapy and use of oral treatments is increasing. We discuss the benefits and challenges of oral chemotherapy from the perspectives of patients, healthcare providers and healthcare funders. Important issues include patient preference, efficacy, compliance, bioavailability, reimbursement, use in special patient populations, financial and staff time savings and flexibility of dosing. We review data for traditional oral agents (e.g. cyclophosphamide, methotrexate), newer oral chemotherapies (e.g. capecitabine), oral formulations of traditionally intravenous agents (e.g. vinorelbine, idarubicin) and new biologic agents under evaluation in breast cancer (e.g. tyrosine kinase inhibitors). Lastly, we review studies of all-oral combination regimens. The wealth of data available and the increasing use of oral agents in breast cancer suggest that many of the concerns and perceptions about oral therapy, including efficacy and bioavailability, have been overcome, and that oral therapy will play a major role in breast cancer management in the future in both the metastatic and adjuvant settings.
Collapse
Affiliation(s)
- M Findlay
- Faculty of Medical & Health Sciences, University of Auckland, Auckland, New Zealand.
| | | | | |
Collapse
|
12
|
Current awareness: Pharmacoepidemiology and drug safety. Pharmacoepidemiol Drug Saf 2007. [DOI: 10.1002/pds.1374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|