1
|
Hu L, Pan X, Hu J, Zeng H, Liu X, Jiang M, Jiang B. Proteasome inhibitors decrease paclitaxel‑induced cell death in nasopharyngeal carcinoma with the accumulation of CDK1/cyclin B1. Int J Mol Med 2021; 48:193. [PMID: 34435645 PMCID: PMC8416144 DOI: 10.3892/ijmm.2021.5026] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 07/02/2021] [Indexed: 12/12/2022] Open
Abstract
Southeast Asia is a region with high incidence of nasopharyngeal carcinoma (NPC). Paclitaxel is the mainstay for the treatment of advanced nasopharyngeal cancer. The present study investigated the effect of proteasome inhibitors on the therapeutic effect of paclitaxel and its related mechanism. The present data from Cell Counting Kit-8 and flow cytometry assays demonstrated that appropriate concentrations of proteasome inhibitors (30 nM PS341 or 700 nM MG132) reduced the lethal effect of paclitaxel on the nasopharyngeal cancer cells. While 400 nM paclitaxel effectively inhibited cell division and induced cell death, proteasome inhibitors (PS341 30 nM or MG132 700 nM) could reverse these effects. Additionally, the western blotting results demonstrated accumulation of cell cycle regulation protein CDK1 and cyclin B1 in proteasome inhibitor-treated cells. In addition, proteasome inhibitors combined with paclitaxel led to decreased MCL1 apoptosis regulator, BCL2 family member/Caspase-9/poly (ADP-ribose) polymerase apoptosis signaling triggered by CDK1/cyclin B1. Therefore, dysfunction of CDK1/cyclin B1 could be defining the loss of paclitaxel lethality against cancer cells, a phenomenon affirmed by the CDK1 inhibitor Ro3306. Overall, the present results demonstrated that a combination of paclitaxel with proteasome inhibitors or CDK1 inhibitors is antagonistic to effective clinical management of NPC.
Collapse
Affiliation(s)
- Ling Hu
- Medical Research Center, Changsha Central Hospital, University of South China, Changsha, Hunan 410004, P.R. China
| | - Xi Pan
- Department of Oncology, Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, P.R. China
| | - Jinyue Hu
- Medical Research Center, Changsha Central Hospital, University of South China, Changsha, Hunan 410004, P.R. China
| | - Hong Zeng
- Reproductive Medicine Center, Foshan Maternal and Child Health Care Hospital, Southern Medical University, Foshan, Guangdong 528000, P.R. China
| | - Xueting Liu
- Medical Research Center, Changsha Central Hospital, University of South China, Changsha, Hunan 410004, P.R. China
| | - Manli Jiang
- Medical Research Center, Changsha Central Hospital, University of South China, Changsha, Hunan 410004, P.R. China
| | - Binyuan Jiang
- Medical Research Center, Changsha Central Hospital, University of South China, Changsha, Hunan 410004, P.R. China
| |
Collapse
|
2
|
Gondal MN, Butt RN, Shah OS, Sultan MU, Mustafa G, Nasir Z, Hussain R, Khawar H, Qazi R, Tariq M, Faisal A, Chaudhary SU. A Personalized Therapeutics Approach Using an In Silico Drosophila Patient Model Reveals Optimal Chemo- and Targeted Therapy Combinations for Colorectal Cancer. Front Oncol 2021; 11:692592. [PMID: 34336681 PMCID: PMC8323493 DOI: 10.3389/fonc.2021.692592] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Accepted: 06/30/2021] [Indexed: 12/18/2022] Open
Abstract
In silico models of biomolecular regulation in cancer, annotated with patient-specific gene expression data, can aid in the development of novel personalized cancer therapeutic strategies. Drosophila melanogaster is a well-established animal model that is increasingly being employed to evaluate such preclinical personalized cancer therapies. Here, we report five Boolean network models of biomolecular regulation in cells lining the Drosophila midgut epithelium and annotate them with colorectal cancer patient-specific mutation data to develop an in silico Drosophila Patient Model (DPM). We employed cell-type-specific RNA-seq gene expression data from the FlyGut-seq database to annotate and then validate these networks. Next, we developed three literature-based colorectal cancer case studies to evaluate cell fate outcomes from the model. Results obtained from analyses of the proposed DPM help: (i) elucidate cell fate evolution in colorectal tumorigenesis, (ii) validate cytotoxicity of nine FDA-approved CRC drugs, and (iii) devise optimal personalized treatment combinations. The personalized network models helped identify synergistic combinations of paclitaxel-regorafenib, paclitaxel-bortezomib, docetaxel-bortezomib, and paclitaxel-imatinib for treating different colorectal cancer patients. Follow-on therapeutic screening of six colorectal cancer patients from cBioPortal using this drug combination demonstrated a 100% increase in apoptosis and a 100% decrease in proliferation. In conclusion, this work outlines a novel roadmap for decoding colorectal tumorigenesis along with the development of personalized combinatorial therapeutics for preclinical translational studies.
Collapse
Affiliation(s)
- Mahnoor Naseer Gondal
- Biomedical Informatics Research Laboratory, Department of Biology, Lahore University of Management Sciences, Lahore, Pakistan
| | - Rida Nasir Butt
- Biomedical Informatics Research Laboratory, Department of Biology, Lahore University of Management Sciences, Lahore, Pakistan
| | - Osama Shiraz Shah
- Biomedical Informatics Research Laboratory, Department of Biology, Lahore University of Management Sciences, Lahore, Pakistan
| | - Muhammad Umer Sultan
- Biomedical Informatics Research Laboratory, Department of Biology, Lahore University of Management Sciences, Lahore, Pakistan
| | - Ghulam Mustafa
- Biomedical Informatics Research Laboratory, Department of Biology, Lahore University of Management Sciences, Lahore, Pakistan
| | - Zainab Nasir
- Biomedical Informatics Research Laboratory, Department of Biology, Lahore University of Management Sciences, Lahore, Pakistan
| | - Risham Hussain
- Biomedical Informatics Research Laboratory, Department of Biology, Lahore University of Management Sciences, Lahore, Pakistan
| | - Huma Khawar
- Biomedical Informatics Research Laboratory, Department of Biology, Lahore University of Management Sciences, Lahore, Pakistan
| | - Romena Qazi
- Department of Pathology, Shaukat Khanum Memorial Cancer Hospital and Research Centre, Lahore, Pakistan
| | - Muhammad Tariq
- Epigenetics Laboratory, Department of Biology, Lahore University of Management Sciences, Lahore, Pakistan
| | - Amir Faisal
- Cancer Therapeutics Laboratory, Department of Biology, Lahore University of Management Sciences, Lahore, Pakistan
| | - Safee Ullah Chaudhary
- Biomedical Informatics Research Laboratory, Department of Biology, Lahore University of Management Sciences, Lahore, Pakistan
| |
Collapse
|
3
|
Lee KA, Cioni M, Robson A, Bataille V. Metastatic porocarcinoma achieving complete radiological and clinical response with pembrolizumab. BMJ Case Rep 2019; 12:12/9/e228917. [PMID: 31492726 DOI: 10.1136/bcr-2018-228917] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
A 67-year-old woman presented in 2012 with a crusty nodule on the left lower limb. Histopathological examination at this time reported a poorly differentiated squamous cell carcinoma (SCC). Two years later, she underwent lymphadenectomy and radiotherapy due to unilateral inguinal and pelvic sidewall nodal metastases. The following year she required excision of two subcutaneous lesions, reported pathologically to be SCC metastases. Further imaging following cyberknife radiotherapy to new brain metastases demonstrated widespread metastatic visceral disease. Twelve cycles of carboplatin and capecitabine failed to halt disease progression. In February 2017, she commenced pembrolizumab, achieving an excellent response and currently has no clinical or radiological evidence of disease. Given the unusual behaviour of her cancer, a histopathological review was requested. The diagnosis was revised to that of porocarcinoma (PC). This represents the first documented case of PC treated with immunotherapy. As of March 2019, the patient remains free of disease.
Collapse
Affiliation(s)
- Karla A Lee
- Department of Twin Research, King's College London, London, UK.,The Royal Marsden NHS Foundation Trust, London, UK
| | - Margherita Cioni
- Department of Dermatology, San Martino Policlinic Hospital, Genoa, Italy
| | - Alistair Robson
- LD Path Group, London, UK.,Department of Pathology, Instituto Portugues de Oncologia de Lisboa Francisco Gentil EPE, Lisboa, Portugal
| | - Veronique Bataille
- Kings College, London, UK.,Dermatology, West Herts NHS Trust, Hemel Hempstead, UK
| |
Collapse
|
4
|
Kabacaoglu D, Ruess DA, Ai J, Algül H. NF-κB/Rel Transcription Factors in Pancreatic Cancer: Focusing on RelA, c-Rel, and RelB. Cancers (Basel) 2019; 11:E937. [PMID: 31277415 PMCID: PMC6679104 DOI: 10.3390/cancers11070937] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2019] [Revised: 06/26/2019] [Accepted: 07/02/2019] [Indexed: 02/07/2023] Open
Abstract
Regulation of Nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB)/Rel transcription factors (TFs) is extremely cell-type-specific owing to their ability to act disparately in the context of cellular homeostasis driven by cellular fate and the microenvironment. This is also valid for tumor cells in which every single component shows heterogenic effects. Whereas many studies highlighted a per se oncogenic function for NF-κB/Rel TFs across cancers, recent advances in the field revealed their additional tumor-suppressive nature. Specifically, pancreatic ductal adenocarcinoma (PDAC), as one of the deadliest malignant diseases, shows aberrant canonical-noncanonical NF-κB signaling activity. Although decades of work suggest a prominent oncogenic activity of NF-κB signaling in PDAC, emerging evidence points to the opposite including anti-tumor effects. Considering the dual nature of NF-κB signaling and how it is closely linked to many other cancer related signaling pathways, it is essential to dissect the roles of individual Rel TFs in pancreatic carcinogenesis and tumor persistency and progression. Here, we discuss recent knowledge highlighting the role of Rel TFs RelA, RelB, and c-Rel in PDAC development and maintenance. Next to providing rationales for therapeutically harnessing Rel TF function in PDAC, we compile strategies currently in (pre-)clinical evaluation.
Collapse
Affiliation(s)
- Derya Kabacaoglu
- Internal Medicine II, Klinikum rechts der Isar, Technische Universität München, 81675 Munich, Germany
| | - Dietrich A Ruess
- Department of Surgery, Faculty of Medicine, Medical Center, University of Freiburg, 79106 Freiburg, Germany
| | - Jiaoyu Ai
- Internal Medicine II, Klinikum rechts der Isar, Technische Universität München, 81675 Munich, Germany
| | - Hana Algül
- Internal Medicine II, Klinikum rechts der Isar, Technische Universität München, 81675 Munich, Germany.
| |
Collapse
|
5
|
Lee SM, Backenroth D, Cheung YKK, Hershman DL, Vulih D, Anderson B, Ivy P, Minasian L. Case Example of Dose Optimization Using Data From Bortezomib Dose-Finding Clinical Trials. J Clin Oncol 2016; 34:1395-401. [PMID: 26926682 DOI: 10.1200/jco.2015.66.0662] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
PURPOSE The current dose-finding methodology for estimating the maximum tolerated dose of investigational anticancer agents is based on the cytotoxic chemotherapy paradigm. Molecularly targeted agents (MTAs) have different toxicity profiles, which may lead to more long-lasting mild or moderate toxicities as well as to late-onset and cumulative toxicities. Several approved MTAs have been poorly tolerated during long-term administration, leading to postmarketing dose optimization studies to re-evaluate the optimal treatment dose. Using data from completed bortezomib dose-finding trials, we explore its toxicity profile, optimize its dose, and examine the appropriateness of current designs for identifying an optimal dose. PATIENTS AND METHODS We classified the toxicities captured from 481 patients in 14 bortezomib dose-finding studies conducted through the National Cancer Institute Cancer Therapy Evaluation Program, computed the incidence of late-onset toxicities, and compared the incidence of dose-limiting toxicities (DLTs) among groups of patients receiving different doses of bortezomib. RESULTS A total of 13,008 toxicities were captured: 46% of patients' first DLTs and 88% of dose reductions or discontinuations of treatment because of toxicity were observed after the first cycle. Moreover, for the approved dose of 1.3 mg/m(2), the estimated cumulative incidence of DLT was > 50%, and the estimated cumulative incidence of dose reduction or treatment discontinuation because of toxicity was nearly 40%. CONCLUSIONS When considering the entire course of treatment, the approved bortezomib dose exceeds the conventional ceiling DLT rate of 20% to 33%. Retrospective analysis of trial data provides an opportunity for dose optimization of MTAs. Future dose-finding studies of MTAs should take into account late-onset toxicities to ensure that a tolerable dose is identified for future efficacy and comparative trials.
Collapse
Affiliation(s)
- Shing M Lee
- Shing M. Lee, Daniel Backenroth, Ying Kuen Ken Cheung, and Dawn L. Hershman, Columbia University, New York, NY; Diana Vulih and Barry Anderson, Theradex Systems, Princeton, NJ; and Percy Ivy and Lori Minasian, National Cancer Institute, Bethesda, MD.
| | - Daniel Backenroth
- Shing M. Lee, Daniel Backenroth, Ying Kuen Ken Cheung, and Dawn L. Hershman, Columbia University, New York, NY; Diana Vulih and Barry Anderson, Theradex Systems, Princeton, NJ; and Percy Ivy and Lori Minasian, National Cancer Institute, Bethesda, MD
| | - Ying Kuen Ken Cheung
- Shing M. Lee, Daniel Backenroth, Ying Kuen Ken Cheung, and Dawn L. Hershman, Columbia University, New York, NY; Diana Vulih and Barry Anderson, Theradex Systems, Princeton, NJ; and Percy Ivy and Lori Minasian, National Cancer Institute, Bethesda, MD
| | - Dawn L Hershman
- Shing M. Lee, Daniel Backenroth, Ying Kuen Ken Cheung, and Dawn L. Hershman, Columbia University, New York, NY; Diana Vulih and Barry Anderson, Theradex Systems, Princeton, NJ; and Percy Ivy and Lori Minasian, National Cancer Institute, Bethesda, MD
| | - Diana Vulih
- Shing M. Lee, Daniel Backenroth, Ying Kuen Ken Cheung, and Dawn L. Hershman, Columbia University, New York, NY; Diana Vulih and Barry Anderson, Theradex Systems, Princeton, NJ; and Percy Ivy and Lori Minasian, National Cancer Institute, Bethesda, MD
| | - Barry Anderson
- Shing M. Lee, Daniel Backenroth, Ying Kuen Ken Cheung, and Dawn L. Hershman, Columbia University, New York, NY; Diana Vulih and Barry Anderson, Theradex Systems, Princeton, NJ; and Percy Ivy and Lori Minasian, National Cancer Institute, Bethesda, MD
| | - Percy Ivy
- Shing M. Lee, Daniel Backenroth, Ying Kuen Ken Cheung, and Dawn L. Hershman, Columbia University, New York, NY; Diana Vulih and Barry Anderson, Theradex Systems, Princeton, NJ; and Percy Ivy and Lori Minasian, National Cancer Institute, Bethesda, MD
| | - Lori Minasian
- Shing M. Lee, Daniel Backenroth, Ying Kuen Ken Cheung, and Dawn L. Hershman, Columbia University, New York, NY; Diana Vulih and Barry Anderson, Theradex Systems, Princeton, NJ; and Percy Ivy and Lori Minasian, National Cancer Institute, Bethesda, MD
| |
Collapse
|
6
|
Uwagawa T, Yanaga K. Effect of NF-κB inhibition on chemoresistance in biliary-pancreatic cancer. Surg Today 2015; 45:1481-8. [PMID: 25673034 DOI: 10.1007/s00595-015-1129-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Accepted: 01/26/2015] [Indexed: 12/13/2022]
Abstract
Biliary cancer and pancreatic cancer are considered to be difficult diseases to cure. Although complete resection provides the only means of curing these cancers, the rate of resectability is not high. Therefore, chemotherapy is often selected in patients with advanced unresectable biliary-pancreatic cancer. Many combination chemotherapy regimens have been applied in clinical trials. However, the survival time is not satisfactory. On the other hand, most chemotherapeutic agents induce anti-apoptotic transcriptional factor nuclear factor kappa b (NF-κB) activation, and agent-induced NF-κB activation is deeply involved in the onset of chemoresistance. Recently, novel approaches to potentiating chemosensitivity in cases of biliary-pancreatic cancer using NF-κB inhibitors with cytotoxic agents have been reported, most of which comprise translational research, although some clinical trials have also been conducted. Nevertheless, to date, there is no breakthrough chemotherapy regimen for these diseases. As some reports show promising data, combination chemotherapy consisting of a NF-κB inhibitor with chemotherapeutic agents seems to improve chemosensitivity and prolong the survival time of biliary-pancreatic cancer patients.
Collapse
Affiliation(s)
- Tadashi Uwagawa
- Department of Surgery, The Jikei University School of Medicine, Tokyo, Japan.
| | - Katsuhiko Yanaga
- Department of Surgery, The Jikei University School of Medicine, Tokyo, Japan
| |
Collapse
|
7
|
Zhang R, Liu Y, Yang Z, Li Y, Rong X, Wang L, Guo C, Li S, Liu J, Li M, Wu Y. Construction of nanoparticles based on amphiphilic PEI–PA polymers for bortezomib and paclitaxel co-delivery. RSC Adv 2015. [DOI: 10.1039/c4ra16544f] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Polymer nanoparticles based on branched polyethyleneimine and palmitic acid conjugates were fabricated for bortezomib and paclitaxel co-delivery.
Collapse
|
8
|
Phase II study of gemcitabine in combination with regional arterial infusion of nafamostat mesilate for advanced pancreatic cancer. Am J Clin Oncol 2013; 36:44-8. [PMID: 22157216 DOI: 10.1097/coc.0b013e31823a53b2] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
PURPOSE To evaluate the efficacy of regional arterial infusion of the synthetic serine protease inhibitor nafamostat mesilate combined with gemcitabine for the treatment of patients with unresectable locally advanced or metastatic pancreatic cancer. MATERIALS AND METHODS A single-arm, single center, institutional review board-approved phase II trial was conducted. Thirty-five of 38 consecutive patients were included in the study. Patients received nafamostat mesilate (4.8 mg/kg continuous regional arterial infusion) with gemcitabine (1000 mg/m intravenously) on days 1, 8, and 15. This treatment was repeated at 28-day intervals. The primary endpoints were to evaluate overall survival and 1-year survival rate. The secondary endpoints were to assess therapeutic response and clinical benefit response. Overall survival times were estimated by the Kaplan-Meier survival analysis. RESULTS The median survival time was 10.0 months, and the 1-year survival rate was 40.0%. The response rate and disease control rate were 17.1% and 88.6%, respectively. A fraction of 25% of the patients who required opioids for cancer-related pain could reduce their opioid intake, and 37.1% of the patients showed healthy weight gain. Among the patients with metastatic pancreatic cancer, the median survival time was 9.0 months, and the 1-year survival rate was 32.0%. The proposed regimen offers an economic advantage compared with recent therapy regimens that have shown significant improvements in median survival over standard chemotherapy with gemcitabine. CONCLUSIONS An alternative regimen for unresectable pancreatic cancer, especially for metastatic pancreatic cancer, is proposed based on acceptable survival time, clinical benefit, and cost advantage.
Collapse
|
9
|
Arlt A, Schäfer H, Kalthoff H. The 'N-factors' in pancreatic cancer: functional relevance of NF-κB, NFAT and Nrf2 in pancreatic cancer. Oncogenesis 2012; 1:e35. [PMID: 23552468 PMCID: PMC3511680 DOI: 10.1038/oncsis.2012.35] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2012] [Accepted: 10/06/2012] [Indexed: 12/12/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) represents one of the deadliest malignancies, with an overall life expectancy of 6 months. Despite considerable advances in the understanding of the molecular mechanisms involved in the carcinogenesis of PDAC, the outcome of the disease was not significantly improved over the last 20 years. Although some achievements in molecular-targeted therapies have been made (that is, targeting the epidermal growth factor receptor by erlotinib), which already entered clinical settings, and despite the promising outcome of the FOLFIRINOX trial, there is an urgent need for improvement of the chemotherapy in this disease. A plethora of molecular alterations are thought to be responsible for the profound chemoresistance, including mutations in oncogenes and tumor suppressors. Besides these classical hallmarks of cancer, the constitutive or inducible activity of transcription factor pathways are characteristic changes in PDAC. Recently, three transcription factors-nuclear factor-κB (NF-κB), nuclear factor of activated T cells (NFAT) and nuclear factor-E2-related factor-2 (Nrf2)-have been shown to be crucial for tumor development and chemoresistance in pancreatic cancer. These transcription factors are key regulators of a variety of genes involved in nearly all aspects of tumorigenesis and resistance against chemotherapeutics and death receptor ligands. Furthermore, the pathways of NF-κB, NFAT and Nrf2 are functional, interacting on several regulatory steps, and, especially, natural compounds such as curcumin interfere with more than one pathway. Thus, targeting these pathways by established inhibitors or new drugs might have great potential to improve the outcome of PDAC patients, most likely in combination with established anticancer drugs. In this article, we summarize recent progress in the characterization of these transcription-factor pathways and their role in PDAC and therapy resistance. We also discuss future concepts for the treatment of PDAC relying on these pathways.
Collapse
Affiliation(s)
- A Arlt
- Laboratory of Molecular Gastroenterology and Hepatology, Department of Internal Medicine I, Kiel, Germany
| | - H Schäfer
- Laboratory of Molecular Gastroenterology and Hepatology, Department of Internal Medicine I, Kiel, Germany
| | - H Kalthoff
- Division of Molecular Oncology, Institute for Experimental Cancer Research, Comprehensive Cancer Center North, Kiel, Germany
| |
Collapse
|
10
|
Mehnert JM, Tan AR, Moss R, Poplin E, Stein MN, Sovak M, Levinson K, Lin H, Kane M, Gounder M, Lin Y, Shih WJ, White E, Rubin EH, Karantza V. Rationally designed treatment for solid tumors with MAPK pathway activation: a phase I study of paclitaxel and bortezomib using an adaptive dose-finding approach. Mol Cancer Ther 2011; 10:1509-19. [PMID: 21680752 PMCID: PMC3155243 DOI: 10.1158/1535-7163.mct-10-0944] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
In the preclinical setting, phosphorylation and subsequent proteosomal degradation of the proapoptotic protein BIM confers resistance to paclitaxel in solid tumors with RAS/RAF/MAPK pathway activation. Concurrent administration of the proteasome inhibitor bortezomib enables paclitaxel-induced BIM accumulation, restoring cancer cell apoptosis in vitro and producing tumor regression in mice in vivo. A phase I study was conducted to determine the maximum tolerated dose (MTD) of paclitaxel and bortezomib combinatorial treatment. Sixteen patients with refractory solid tumors commonly exhibiting mitogen-activated protein kinase (MAPK) pathway activation were treated weekly with paclitaxel and bortezomib. Starting doses were 40 mg/m(2) for paclitaxel and 0.7 mg/m(2) for bortezomib. A modified continual reassessment method adapted for 2-drug escalation was used for MTD determination with 3-patient cohorts treated at each dose level. MTD was reached at 60 mg/m(2) paclitaxel and 1.0 mg/m(2) bortezomib, the recommended phase II dose. Therapy was overall well tolerated. Most frequently observed toxicities included anemia (in 43.75% of patients, one grade 3 event), fatigue (in 43.75% of patients, one grade 3 event beyond cycle 1), and neuropathy (in 31.25% of patients, one grade 3 event after cycle 1). Of 15 evaluable patients, one non-small-cell lung carcinoma (NSCLC) patient with paclitaxel exposure at the adjuvant setting had a partial response and five patients had stable disease (SD); median disease stabilization was 143.5 days; three NSCLC patients had SD lasting 165 days or longer. Thus, rationally designed weekly treatment with paclitaxel and bortezomib in solid tumors with MAPK pathway activation, including previously taxane-treated malignancies, is a tolerable regimen with preliminary signals of antitumor activity worthy of further investigation.
Collapse
Affiliation(s)
- Janice M. Mehnert
- Department of Medicine, Division of Medical Oncology, University of Medicine and Dentistry of New Jersey, Robert Wood Johnson Medical School, Piscataway, NJ
- The Cancer Institute of New Jersey, New Brunswick, NJ
| | - Antoinette R. Tan
- Department of Medicine, Division of Medical Oncology, University of Medicine and Dentistry of New Jersey, Robert Wood Johnson Medical School, Piscataway, NJ
- The Cancer Institute of New Jersey, New Brunswick, NJ
| | - Rebecca Moss
- Department of Medicine, Division of Medical Oncology, University of Medicine and Dentistry of New Jersey, Robert Wood Johnson Medical School, Piscataway, NJ
- The Cancer Institute of New Jersey, New Brunswick, NJ
| | - Elizabeth Poplin
- Department of Medicine, Division of Medical Oncology, University of Medicine and Dentistry of New Jersey, Robert Wood Johnson Medical School, Piscataway, NJ
- The Cancer Institute of New Jersey, New Brunswick, NJ
| | - Mark N. Stein
- Department of Medicine, Division of Medical Oncology, University of Medicine and Dentistry of New Jersey, Robert Wood Johnson Medical School, Piscataway, NJ
- The Cancer Institute of New Jersey, New Brunswick, NJ
| | - Mika Sovak
- Department of Medicine, Division of Medical Oncology, University of Medicine and Dentistry of New Jersey, Robert Wood Johnson Medical School, Piscataway, NJ
- The Cancer Institute of New Jersey, New Brunswick, NJ
| | | | - Hongxia Lin
- The Cancer Institute of New Jersey, New Brunswick, NJ
| | - Michael Kane
- Pharmacy, The Cancer Institute of New Jersey, New Brunswick, NJ
| | - Murugesan Gounder
- The Cancer Institute of New Jersey, New Brunswick, NJ
- Department of Pharmacology, University of Medicine and Dentistry of New Jersey, Robert Wood Johnson Medical School, Piscataway, NJ
| | - Yong Lin
- The Cancer Institute of New Jersey, New Brunswick, NJ
- Department of Biostatistics, University of Medicine and Dentistry of New Jersey, School of Public Health, Piscataway, NJ
| | - Weichung Joe Shih
- The Cancer Institute of New Jersey, New Brunswick, NJ
- Department of Biostatistics, University of Medicine and Dentistry of New Jersey, School of Public Health, Piscataway, NJ
| | - Eileen White
- The Cancer Institute of New Jersey, New Brunswick, NJ
- Department of Molecular Biology and Biochemistry, Rutgers University, Piscataway, NJ
| | - Eric H. Rubin
- Department of Medicine, Division of Medical Oncology, University of Medicine and Dentistry of New Jersey, Robert Wood Johnson Medical School, Piscataway, NJ
- The Cancer Institute of New Jersey, New Brunswick, NJ
- Department of Pharmacology, University of Medicine and Dentistry of New Jersey, Robert Wood Johnson Medical School, Piscataway, NJ
| | - Vassiliki Karantza
- Department of Medicine, Division of Medical Oncology, University of Medicine and Dentistry of New Jersey, Robert Wood Johnson Medical School, Piscataway, NJ
- The Cancer Institute of New Jersey, New Brunswick, NJ
| |
Collapse
|