101
|
Current controversies in the management of metastatic colorectal cancer. Cancer Chemother Pharmacol 2015; 76:659-77. [DOI: 10.1007/s00280-015-2808-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Accepted: 06/12/2015] [Indexed: 12/16/2022]
|
102
|
Rihawi K, Giampieri R, Scartozzi M, Pusceddu V, Bonotto M, Fasola G, Cascinu S, Aprile G. Role and mechanisms of resistance of epidermal growth factor receptor antagonists in the treatment of colorectal cancer. Expert Opin Investig Drugs 2015; 24:1185-98. [DOI: 10.1517/13543784.2015.1054479] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
103
|
Scartozzi M, Giampieri R, Aprile G, Iacono D, Santini D, dell’Aquila E, Silvestris N, Gnoni A, Bonotto M, Puzzoni M, Demurtas L, Cascinu S. The distinctive molecular, pathological and clinical characteristics ofBRAF-mutant colorectal tumors. Expert Rev Mol Diagn 2015; 15:979-87. [DOI: 10.1586/14737159.2015.1047346] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
|
104
|
|
105
|
Abstract
Colorectal cancer is a serious health problem, a challenge for research, and a model for studying the molecular mechanisms involved in its development. According to its incidence, this pathology manifests itself in three forms: family, hereditary, and most commonly sporadic, apparently not associated with any hereditary or familial factor. For the types having inheritance patterns and a family predisposition, the tumours develop through defined stages ranging from adenomatous lesions to the manifestation of a malignant tumour. It has been established that environmental and hereditary factors contribute to the development of colorectal cancer, as indicated by the accumulation of mutations in oncogenes, genes which suppress and repair DNA, signaling the existence of various pathways through which the appearance of tumours may occur. In the case of the suppressive and mutating tracks, these are characterised by genetic disorders related to the phenotypical changes of the morphological progression sequence in the adenoma/carcinoma. Moreover, alternate pathways through mutation in BRAF and KRAS genes are associated with the progression of polyps to cancer. This review surveys the research done at the cellular and molecular level aimed at finding specific alternative therapeutic targets for fighting colorectal cancer.
Collapse
Affiliation(s)
- Francisco Arvelo
- Centre for Biosciences, Institute for Advanced Studies Foundation-IDEA, Caracas 1015-A, Apartado 17606, Venezuela ; Laboratory for Tissue Culture and Tumour Biology, Institute of Experimental Biology, Central University of Venezuela, Apartado 47114, Caracas, Venezuela
| | - Felipe Sojo
- Centre for Biosciences, Institute for Advanced Studies Foundation-IDEA, Caracas 1015-A, Apartado 17606, Venezuela ; Laboratory for Tissue Culture and Tumour Biology, Institute of Experimental Biology, Central University of Venezuela, Apartado 47114, Caracas, Venezuela
| | - Carlos Cotte
- Laboratory for Tissue Culture and Tumour Biology, Institute of Experimental Biology, Central University of Venezuela, Apartado 47114, Caracas, Venezuela
| |
Collapse
|
106
|
Cross-resistance to clinically used tyrosine kinase inhibitors sunitinib, sorafenib and pazopanib. Cell Oncol (Dordr) 2015; 38:119-29. [PMID: 25665527 PMCID: PMC4555235 DOI: 10.1007/s13402-015-0218-8] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/13/2015] [Indexed: 01/24/2023] Open
Abstract
PURPOSE When during cancer treatment resistance to a tyrosine kinase inhibitor (TKI) occurs, switching to another TKI is often considered as a reasonable option. Previously, we reported that resistance to sunitinib may be caused by increased lysosomal sequestration, leading to increased intracellular lysosomal storage and, thereby, inactivity. Here, we studied the effect of several other TKIs on the development of (cross-) resistance. METHODS TKI resistance was induced by continuous exposure of cancer cell lines to increasing TKI concentrations for 3-4 months. (Cross-) resistance was evaluated using MTT cell proliferation assays. Intracellular TKI concentrations were measured using LC-MS/MS. Western blotting was used to detect lysosome-associated membrane protein-1 and -2 (LAMP1/2) expression. RESULTS The previously generated sunitinib-resistant (SUN) renal cancer cells (786-O) and colorectal cancer cells (HT-29) were found to be cross-resistant to pazopanib, erlotinib and lapatinib, but not sorafenib. Exposure of 786-O and HT-29 cells to sorafenib, pazopanib or erlotinib for 3-4 months induced drug resistance to pazopanib and erlotinib, but not sorafenib. Intracellular drug accumulation was found to be increased in pazopanib- and erlotinib-, but not in sorafenib-exposed cells. Lysosomal capacity, reflected by LAMP1/2 expression, was found to be increased in resistant cells and, in addition, to be transient. No cross-resistance to the mTOR inhibitor everolimus was detected. CONCLUSIONS Our data indicate that tumor cells can develop (cross-) resistance to TKIs, and that such resistance includes increased intracellular drug accumulation accompanied by increased lysosomal storage. Transient (cross-) resistance was found to occur for several of the TKIs tested, but not for everolimus, indicating that switching from a TKI to a mTOR inhibitor may be an attractive therapeutic option.
Collapse
|
107
|
Akkad J, Bochum S, Martens UM. Personalized treatment for colorectal cancer: novel developments and putative therapeutic strategies. Langenbecks Arch Surg 2015; 400:129-43. [DOI: 10.1007/s00423-015-1276-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Accepted: 01/23/2015] [Indexed: 01/14/2023]
|
108
|
Pal S, Konkimalla VB, Kathawate L, Rao SS, Gejji SP, Puranik VG, Weyhermüller T, Salunke-Gawali S. Targeting a chemorefractory COLO205 (BRAF V600E) cell line using substituted benzo[α]phenoxazines. RSC Adv 2015. [DOI: 10.1039/c5ra14949e] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Benzo[α]phenoxazine derivatives specifically toxic to a malignant COLO205 cell line with a BRAF mutation (V600E) and nontoxic to a non-malignant wild-type BRAF HEK293T cell line are studied.
Collapse
Affiliation(s)
- Sanjima Pal
- School of Biological Sciences
- National Institute of Science Education and Research (NISER)
- Bhubaneswar 751005
- India
| | - V. Badireenath Konkimalla
- School of Biological Sciences
- National Institute of Science Education and Research (NISER)
- Bhubaneswar 751005
- India
| | - Laxmi Kathawate
- Department of Chemistry
- Savitribai Phule Pune University
- Pune 411007
- India
| | - Soniya S. Rao
- Department of Chemistry
- Savitribai Phule Pune University
- Pune 411007
- India
| | - Shridhar P. Gejji
- Department of Chemistry
- Savitribai Phule Pune University
- Pune 411007
- India
| | - Vedavati G. Puranik
- Center for Material Characterization
- National Chemical Laboratory
- Pune 411008
- India
| | | | | |
Collapse
|
109
|
Santhosh S, Kumar P, Ramprasad V, Chaudhuri A. Evolution of targeted therapies in cancer: opportunities and challenges in the clinic. Future Oncol 2015; 11:279-93. [DOI: 10.2217/fon.14.198] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
ABSTRACT Targeted therapies have changed the course of cancer treatment in recent years. By reducing toxicity and improving outcome, these new generations of precision medicines have extended patient lives beyond what could be achieved by the use of nontargeted therapies. In the last 2 years, several new molecular entities targeting signaling proteins and immune pathways have gone through successful clinical development resulting in their approval. These new targeted therapies require patient selection and the discovery of biomarkers of response. This review discusses the evolution of targeted therapies in cancer and challenges in translating the concepts into clinical practice.
Collapse
|
110
|
Wilson MK, Collyar D, Chingos DT, Friedlander M, Ho TW, Karakasis K, Kaye S, Parmar MKB, Sydes MR, Tannock IF, Oza AM. Outcomes and endpoints in cancer trials: bridging the divide. Lancet Oncol 2015; 16:e43-52. [PMID: 25638556 DOI: 10.1016/s1470-2045(14)70380-8] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Cancer is not one disease. Outcomes and endpoints in trials should incorporate the therapeutic modality and cancer type because these factors affect clinician and patient expectations. In this Review, we discuss how to: define the importance of endpoints; make endpoints understandable to patients; improve the use of patient-reported outcomes; advance endpoints to parallel changes in trial design and therapeutic interventions; and integrate these improvements into trials and practice. Endpoints need to reflect benefit to patients, and show that changes in tumour size either in absolute terms (response and progression) or relative to control (progression) are clinically relevant. Improvements in trial design should be accompanied by improvements in available endpoints. Stakeholders need to come together to determine the best approach for research that ensures accountability and optimises the use of available resources.
Collapse
Affiliation(s)
- Michelle K Wilson
- University of Toronto Princess Margaret Cancer Centre, Toronto, ON, Canada
| | | | | | - Michael Friedlander
- Prince of Wales Clinical School, University of New South Wales, Sydney, Australia
| | - Tony W Ho
- AstraZeneca, Wilmington DE 19850-5437, USA
| | | | - Stan Kaye
- Drug Development Unit and Gynaecology Unit, Royal Marsden Hospital and Institute of Cancer Research, London, UK
| | | | - Matthew R Sydes
- MRC Clinical Trials Unit, University College London, London, UK
| | - Ian F Tannock
- University of Toronto Princess Margaret Cancer Centre, Toronto, ON, Canada
| | - Amit M Oza
- University of Toronto Princess Margaret Cancer Centre, Toronto, ON, Canada.
| |
Collapse
|
111
|
Co-targeting BRAF and cyclin dependent kinases 4/6 for BRAF mutant cancers. Pharmacol Ther 2014; 149:139-49. [PMID: 25550229 DOI: 10.1016/j.pharmthera.2014.12.003] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Accepted: 12/17/2014] [Indexed: 12/19/2022]
Abstract
Selective BRAF inhibitors have demonstrated significant clinical benefit in melanoma patients harboring oncogenic BRAF mutations. However, the majority of such patients either exhibit de novo resistance from the beginning of the treatment or acquire resistance and eventually relapse. Despite tremendous progress in understanding the underlying mechanisms of resistance, overcoming resistance to BRAF inhibitors remains an unmet medical need. Constitutive activation of cyclin-dependent kinases (CDK) 4/6 as a result of genetic aberrations including CDKN2A inactivation and CCND1 amplification is common across many cancer types and frequently co-occurs with oncogenic BRAF mutations. Also, cyclin D1 overexpression is a common feature of resistance to BRAF inhibitors. Here we review CDK4/6 as a therapeutic target in BRAF mutant cancers and discuss emerging evidence supporting a critical role of cyclin D1/CDK4/6 axis in de novo and acquired resistance to BRAF inhibitors. Co-targeting CDK4/6 and BRAF could be a more effective therapy to augment clinical response of BRAF inhibitors and overcome resistance in BRAF mutant cancers.
Collapse
|
112
|
BRAF inhibitors: experience in thyroid cancer and general review of toxicity. Discov Oncol 2014; 6:21-36. [PMID: 25467940 DOI: 10.1007/s12672-014-0207-9] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2014] [Accepted: 11/11/2014] [Indexed: 12/20/2022] Open
Abstract
The US Food and Drug Administration-approved BRAF inhibitors, vemurafenib and dabrafenib, have demonstrated superior efficacy in patients with BRAF-mutant melanomas but have limited efficacy in BRAF-mutant colorectal cancer. Little is known at this time regarding BRAF inhibitors in thyroid cancer. Initial reports in patients with progressive, radioactive iodine-refractory BRAF-mutant papillary thyroid cancer suggest response rates of approximately 30-40%. In this review, we discuss BRAF inhibitors in the context of thyroid cancer, the toxicities associated with BRAF inhibitors, and the suggested management of those toxicities. The management of vemurafenib and dabrafenib toxicities is applicable across all tumor types and may serve as a practical guide to their use.
Collapse
|
113
|
San Lucas FA, Fowler J, Chang K, Kopetz S, Vilar E, Scheet P. Cancer in silico drug discovery: a systems biology tool for identifying candidate drugs to target specific molecular tumor subtypes. Mol Cancer Ther 2014; 13:3230-40. [PMID: 25349306 PMCID: PMC4341901 DOI: 10.1158/1535-7163.mct-14-0260] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Large-scale cancer datasets such as The Cancer Genome Atlas (TCGA) allow researchers to profile tumors based on a wide range of clinical and molecular characteristics. Subsequently, TCGA-derived gene expression profiles can be analyzed with the Connectivity Map (CMap) to find candidate drugs to target tumors with specific clinical phenotypes or molecular characteristics. This represents a powerful computational approach for candidate drug identification, but due to the complexity of TCGA and technology differences between CMap and TCGA experiments, such analyses are challenging to conduct and reproduce. We present Cancer in silico Drug Discovery (CiDD; scheet.org/software), a computational drug discovery platform that addresses these challenges. CiDD integrates data from TCGA, CMap, and Cancer Cell Line Encyclopedia (CCLE) to perform computational drug discovery experiments, generating hypotheses for the following three general problems: (i) determining whether specific clinical phenotypes or molecular characteristics are associated with unique gene expression signatures; (ii) finding candidate drugs to repress these expression signatures; and (iii) identifying cell lines that resemble the tumors being studied for subsequent in vitro experiments. The primary input to CiDD is a clinical or molecular characteristic. The output is a biologically annotated list of candidate drugs and a list of cell lines for in vitro experimentation. We applied CiDD to identify candidate drugs to treat colorectal cancers harboring mutations in BRAF. CiDD identified EGFR and proteasome inhibitors, while proposing five cell lines for in vitro testing. CiDD facilitates phenotype-driven, systematic drug discovery based on clinical and molecular data from TCGA.
Collapse
Affiliation(s)
- F Anthony San Lucas
- The Graduate School of Biomedical Sciences, The University of Texas Health Science Center at Houston, Houston, Texas. Department of Epidemiology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Jerry Fowler
- Department of Epidemiology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Kyle Chang
- The Graduate School of Biomedical Sciences, The University of Texas Health Science Center at Houston, Houston, Texas
| | - Scott Kopetz
- The Graduate School of Biomedical Sciences, The University of Texas Health Science Center at Houston, Houston, Texas. Department of Gastrointestinal Medical Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Eduardo Vilar
- The Graduate School of Biomedical Sciences, The University of Texas Health Science Center at Houston, Houston, Texas. Department of Gastrointestinal Medical Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas. Department of Clinical Cancer Prevention, The University of Texas M.D. Anderson Cancer Center, Houston, Texas.
| | - Paul Scheet
- The Graduate School of Biomedical Sciences, The University of Texas Health Science Center at Houston, Houston, Texas. Department of Epidemiology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas.
| |
Collapse
|
114
|
Theiss AP, Chafin D, Bauer DR, Grogan TM, Baird GS. Immunohistochemistry of colorectal cancer biomarker phosphorylation requires controlled tissue fixation. PLoS One 2014; 9:e113608. [PMID: 25409462 PMCID: PMC4237459 DOI: 10.1371/journal.pone.0113608] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2014] [Accepted: 10/28/2014] [Indexed: 12/31/2022] Open
Abstract
Phosphorylated signaling molecules are biomarkers of cancer pathophysiology and resistance to therapy, but because phosphoprotein analytes are often labile, poorly controlled clinical laboratory practices could prevent translation of research findings in this area from the bench to the bedside. We therefore compared multiple biomarker and phosphoprotein immunohistochemistry (IHC) results in 23 clinical colorectal carcinoma samples after either a novel, rapid tissue fixation protocol or a standard tissue fixation protocol employed by clinical laboratories, and we also investigated the effect of a defined post-operative "cold" ischemia period on these IHC results. We found that a one-hour cold ischemia interval, allowed by ASCO/CAP guidelines for certain cancer biomarker assays, is highly deleterious to certain phosphoprotein analytes, specifically the phosphorylated epidermal growth factor receptor (pEGFR), but shorter ischemic intervals (less than 17 minutes) facilitate preservation of phosphoproteins. Second, we found that a rapid 4-hour, two temperature, formalin fixation yielded superior staining in several cases with select markers (pEGFR, pBAD, pAKT) compared to a standard overnight room temperature fixation protocol, despite taking less time. These findings indicate that the future research and clinical utilities of phosphoprotein IHC for assessing colorectal carcinoma pathophysiology absolutely depend upon attention to preanalytical factors and rigorously controlled tissue fixation protocols.
Collapse
Affiliation(s)
- Abbey P. Theiss
- Ventana Medical Systems Inc., Tucson, Arizona, United States of America
| | - David Chafin
- Ventana Medical Systems Inc., Tucson, Arizona, United States of America
| | - Daniel R. Bauer
- Ventana Medical Systems Inc., Tucson, Arizona, United States of America
| | - Thomas M. Grogan
- Ventana Medical Systems Inc., Tucson, Arizona, United States of America
| | - Geoffrey S. Baird
- Department of Laboratory Medicine, University of Washington, Seattle, Washington, United States of America
- Department of Pathology, University of Washington, Seattle, Washington, United States of America
- * E-mail:
| |
Collapse
|
115
|
Herr R, Köhler M, Andrlová H, Weinberg F, Möller Y, Halbach S, Lutz L, Mastroianni J, Klose M, Bittermann N, Kowar S, Zeiser R, Olayioye MA, Lassmann S, Busch H, Boerries M, Brummer T. B-Raf inhibitors induce epithelial differentiation in BRAF-mutant colorectal cancer cells. Cancer Res 2014; 75:216-29. [PMID: 25381152 DOI: 10.1158/0008-5472.can-13-3686] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
BRAF mutations are associated with aggressive, less-differentiated and therapy-resistant colorectal carcinoma. However, the underlying mechanisms for these correlations remain unknown. To understand how oncogenic B-Raf contributes to carcinogenesis, in particular to aspects other than cellular proliferation and survival, we generated three isogenic human colorectal carcinoma cell line models in which we can dynamically modulate the expression of the B-Raf(V600E) oncoprotein. Doxycyclin-inducible knockdown of endogenous B-Raf(V600E) decreases cellular motility and invasion in conventional and three-dimensional (3D) culture, whereas it promotes cell-cell contacts and induces various hallmarks of differentiated epithelia. Importantly, all these effects are recapitulated by B-Raf (PLX4720, vemurafenib, and dabrafenib) or MEK inhibitors (trametinib). Surprisingly, loss of B-Raf(V600E) in HT29 xenografts does not only stall tumor growth, but also induces glandular structures with marked expression of CDX2, a tumor-suppressor and master transcription factor of intestinal differentiation. By performing the first transcriptome profiles of PLX4720-treated 3D cultures of HT29 and Colo-205 cells, we identify several upregulated genes linked to epithelial differentiation and effector functions, such as claudin-1, a Cdx-2 target gene encoding a critical tight junction component. Thereby, we provide a mechanism for the clinically observed correlation between mutant BRAF and the loss of Cdx-2 and claudin-1. PLX4720 also suppressed several metastasis-associated transcripts that have not been implicated as targets, effectors or potential biomarkers of oncogenic B-Raf signaling so far. Together, we identify a novel facet of clinically applied B-Raf or MEK inhibitors by showing that they promote cellular adhesion and differentiation of colorectal carcinoma cells.
Collapse
Affiliation(s)
- Ricarda Herr
- Signal Transduction in Tumour Development and Drug Resistance Group, Institute of Molecular Medicine and Cell Research (IMMZ), Albert-Ludwigs-University (ALU), Freiburg, Germany. Faculty of Biology, ALU, Freiburg, Germany
| | - Martin Köhler
- Signal Transduction in Tumour Development and Drug Resistance Group, Institute of Molecular Medicine and Cell Research (IMMZ), Albert-Ludwigs-University (ALU), Freiburg, Germany. Faculty of Biology, ALU, Freiburg, Germany. Spemann Graduate School of Biology and Medicine (SGBM), ALU, Freiburg, Germany
| | - Hana Andrlová
- Department of Hematology and Oncology, University Medical Center, ALU, Freiburg, Germany
| | - Florian Weinberg
- Signal Transduction in Tumour Development and Drug Resistance Group, Institute of Molecular Medicine and Cell Research (IMMZ), Albert-Ludwigs-University (ALU), Freiburg, Germany. Faculty of Biology, ALU, Freiburg, Germany
| | - Yvonne Möller
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
| | - Sebastian Halbach
- Signal Transduction in Tumour Development and Drug Resistance Group, Institute of Molecular Medicine and Cell Research (IMMZ), Albert-Ludwigs-University (ALU), Freiburg, Germany. Faculty of Biology, ALU, Freiburg, Germany. Spemann Graduate School of Biology and Medicine (SGBM), ALU, Freiburg, Germany
| | - Lisa Lutz
- Department of Pathology, University Medical Center, ALU, Freiburg, Germany
| | - Justin Mastroianni
- Faculty of Biology, ALU, Freiburg, Germany. Department of Hematology and Oncology, University Medical Center, ALU, Freiburg, Germany
| | - Martin Klose
- Systems Biology of the Cellular Microenvironment Group, IMMZ, ALU, Freiburg, Germany
| | - Nicola Bittermann
- Department of Pathology, University Medical Center, ALU, Freiburg, Germany
| | - Silke Kowar
- Systems Biology of the Cellular Microenvironment Group, IMMZ, ALU, Freiburg, Germany
| | - Robert Zeiser
- Department of Hematology and Oncology, University Medical Center, ALU, Freiburg, Germany. Centre for Biological Signalling Studies BIOSS, ALU Freiburg
| | - Monilola A Olayioye
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
| | - Silke Lassmann
- Department of Pathology, University Medical Center, ALU, Freiburg, Germany. Centre for Biological Signalling Studies BIOSS, ALU Freiburg. German Cancer Consortium (DKTK), Freiburg, Germany. German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Hauke Busch
- Systems Biology of the Cellular Microenvironment Group, IMMZ, ALU, Freiburg, Germany. German Cancer Consortium (DKTK), Freiburg, Germany. German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Melanie Boerries
- Systems Biology of the Cellular Microenvironment Group, IMMZ, ALU, Freiburg, Germany. German Cancer Consortium (DKTK), Freiburg, Germany. German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Tilman Brummer
- Signal Transduction in Tumour Development and Drug Resistance Group, Institute of Molecular Medicine and Cell Research (IMMZ), Albert-Ludwigs-University (ALU), Freiburg, Germany. Centre for Biological Signalling Studies BIOSS, ALU Freiburg.
| |
Collapse
|
116
|
Tie J, Desai J. Targeting BRAF mutant metastatic colorectal cancer: clinical implications and emerging therapeutic strategies. Target Oncol 2014; 10:179-88. [PMID: 25119972 DOI: 10.1007/s11523-014-0330-0] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2014] [Accepted: 07/24/2014] [Indexed: 12/16/2022]
Abstract
Increasing knowledge of the underlying signaling pathways and molecular defects involved in colorectal cancer growth or progression enabled the discovery of several prognostic and predictive biomarkers, leading to the development of novel molecularly targeted therapies. The mitogen-activated protein kinase (MAPK) signaling pathway plays a critical role in colorectal cancer progression. Mutations in BRAF, a principal effector of Ras in this signaling cascade, are found in 10 % of colorectal cancer and play a clear pathogenic role, particularly in patients with metastatic disease. Intense efforts have therefore focused on targeting BRAF as an oncogenic driver, with mixed early results. This article summarizes the molecular and clinical features of BRAF mutant colorectal cancer, the prognostic and predictive role of BRAFV600E mutation in colorectal cancer, initial clinical trial results in targeting BRAFV600E, and the more recent preclinical insights into potential mechanisms of resistance to BRAF inhibition that have now led to a number of rationale-driven combination therapeutic strategies.
Collapse
Affiliation(s)
- Jeanne Tie
- Ludwig Colon Cancer Initiative Laboratory, Ludwig Institute for Cancer Research, Parkville, VIC, Australia
| | | |
Collapse
|
117
|
Jeong WJ, Cha PH, Choi KY. Strategies to overcome resistance to epidermal growth factor receptor monoclonal antibody therapy in metastatic colorectal cancer. World J Gastroenterol 2014; 20:9862-9871. [PMID: 25110417 PMCID: PMC4123368 DOI: 10.3748/wjg.v20.i29.9862] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2013] [Revised: 01/14/2014] [Accepted: 04/16/2014] [Indexed: 02/06/2023] Open
Abstract
Administration of monoclonal antibodies (mAbs) against epidermal growth factor receptor (EGFR) such as cetuximab and panitumumab in combination with conventional chemotherapy substantially prolongs survival of patients with metastatic colorectal cancer (mCRC). However, the efficacy of these mAbs is limited due to genetic variation among patients, in particular K-ras mutations. The discovery of K-ras mutation as a predictor of non-responsiveness to EGFR mAb therapy has caused a major change in the treatment of mCRC. Drugs that inhibit transformation caused by oncogenic alterations of Ras and its downstream components such as BRAF, MEK and AKT seem to be promising cancer therapeutics as single agents or when given with EGFR inhibitors. Although multiple therapeutic strategies to overcome EGFR mAb-resistance are under investigation, our understanding of their mode of action is limited. Rational drug development based on stringent preclinical data, biomarker validation, and proper selection of patients is of paramount importance in the treatment of mCRC. In this review, we will discuss diverse approaches to overcome the problem of resistance to existing anti-EGFR therapies and potential future directions for cancer therapies related to the mutational status of genes associated with EGFR-Ras-ERK and PI3K signalings.
Collapse
|
118
|
Strickler JH, Hurwitz HI. Palliative treatment of metastatic colorectal cancer: what is the optimal approach? Curr Oncol Rep 2014; 16:363. [PMID: 24293074 DOI: 10.1007/s11912-013-0363-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Worldwide, colorectal cancer (CRC) is responsible for over 600,000 deaths annually and remains a significant public health concern. Because of therapeutic advancements over the past two decades, patients with metastatic CRC are living longer with an improved quality of life. This review will highlight recent trial evidence that improves outcomes for patients with metastatic disease. Topics will include the optimal use of first-line combination chemotherapy, bevacizumab in patients with advanced age or comorbidities, maintenance chemotherapy, first-line use of anti-EGFR therapies, first-line cetuximab versus bevacizumab, anti-angiogenic therapies past progression, and management of treatment-refractory disease. Clinical trial evidence will be presented, along with guidance on how to integrate recent evidence into clinical practice. Finally, this review will examine innovative drug development strategies, and will discuss potentially actionable targets identified by molecular testing.
Collapse
Affiliation(s)
- John H Strickler
- Department of Medicine, Division of Medical Oncology, Duke University Medical Center, DUMC 2823, Durham, NC, 27710, USA,
| | | |
Collapse
|
119
|
The B-Raf(V600E) inhibitor dabrafenib selectively inhibits RIP3 and alleviates acetaminophen-induced liver injury. Cell Death Dis 2014; 5:e1278. [PMID: 24901049 PMCID: PMC4611716 DOI: 10.1038/cddis.2014.241] [Citation(s) in RCA: 196] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2014] [Revised: 04/22/2014] [Accepted: 04/23/2014] [Indexed: 12/22/2022]
Abstract
Receptor-interacting protein (RIP)3 is a critical regulator of necroptosis and has been demonstrated to be associated with various diseases, suggesting that its inhibitors are promising in the clinic. However, there have been few RIP3 inhibitors reported as yet. B-RafV600E inhibitors are an important anticancer drug class for metastatic melanoma therapy. In this study, we found that 6 B-Raf inhibitors could inhibit RIP3 enzymatic activity in vitro. Among them, dabrafenib showed the most potent inhibition on RIP3, which was achieved by its ATP-competitive binding to the enzyme. Dabrafenib displayed highly selective inhibition on RIP3 over RIP1, RIP2 and RIP5. Moreover, only dabrafenib rescued cells from RIP3-mediated necroptosis induced by the necroptosis-induced combinations, that is, tumor necrosis factor (TNF)α, TNF-related apoptosis-inducing ligand or Fas ligand plus Smac mimetic and the caspase inhibitor z-VAD. Dabrafenib decreased the RIP3-mediated Ser358 phosphorylation of mixed lineage kinase domain-like protein (MLKL) and disrupted the interaction between RIP3 and MLKL. Notably, RIP3 inhibition of dabrafenib appeared to be independent of its B-Raf inhibition. Dabrafenib was further revealed to prevent acetaminophen-induced necrosis in normal human hepatocytes, which is considered to be mediated by RIP3. In acetaminophen-overdosed mouse models, dabrafenib was found to apparently ease the acetaminophen-caused liver damage. The results indicate that the anticancer B-RafV600E inhibitor dabrafenib is a RIP3 inhibitor, which could serve as a sharp tool for probing the RIP3 biology and as a potential preventive or therapeutic agent for RIP3-involved necroptosis-related diseases such as acetaminophen-induced liver damage.
Collapse
|
120
|
Sweeney RT, McClary AC, Myers BR, Biscocho J, Neahring L, Kwei KA, Qu K, Gong X, Ng T, Jones CD, Varma S, Odegaard JI, Sugiyama T, Koyota S, Rubin BP, Troxell ML, Pelham RJ, Zehnder JL, Beachy PA, Pollack JR, West RB. Identification of recurrent SMO and BRAF mutations in ameloblastomas. Nat Genet 2014; 46:722-5. [PMID: 24859340 DOI: 10.1038/ng.2986] [Citation(s) in RCA: 239] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2013] [Accepted: 04/21/2014] [Indexed: 12/18/2022]
Abstract
Here we report the discovery of oncogenic mutations in the Hedgehog and mitogen-activated protein kinase (MAPK) pathways in over 80% of ameloblastomas, locally destructive odontogenic tumors of the jaw, by genomic analysis of archival material. Mutations in SMO (encoding Smoothened, SMO) are common in ameloblastomas of the maxilla, whereas BRAF mutations are predominant in tumors of the mandible. We show that a frequently occurring SMO alteration encoding p.Leu412Phe is an activating mutation and that its effect on Hedgehog-pathway activity can be inhibited by arsenic trioxide (ATO), an anti-leukemia drug approved by the US Food and Drug Administration (FDA) that is currently in clinical trials for its Hedgehog-inhibitory activity. In a similar manner, ameloblastoma cells harboring an activating BRAF mutation encoding p.Val600Glu are sensitive to the BRAF inhibitor vemurafenib. Our findings establish a new paradigm for the diagnostic classification and treatment of ameloblastomas.
Collapse
Affiliation(s)
- Robert T Sweeney
- 1] Department of Pathology, Stanford University, Stanford, California, USA. [2]
| | - Andrew C McClary
- 1] Department of Pathology, Stanford University, Stanford, California, USA. [2]
| | - Benjamin R Myers
- 1] Department of Biochemistry, Stanford University, Stanford, California, USA. [2] Department of Developmental Biology, Stanford University, Stanford, California, USA. [3] Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, California, USA. [4] Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, California, USA. [5]
| | - Jewison Biscocho
- 1] Department of Pathology, Stanford University, Stanford, California, USA. [2]
| | - Lila Neahring
- 1] Department of Biochemistry, Stanford University, Stanford, California, USA. [2] Department of Developmental Biology, Stanford University, Stanford, California, USA. [3] Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, California, USA. [4] Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, California, USA
| | - Kevin A Kwei
- 1] Genomic Health, Redwood City, California, USA. [2]
| | - Kunbin Qu
- Genomic Health, Redwood City, California, USA
| | - Xue Gong
- Department of Pathology, Stanford University, Stanford, California, USA
| | - Tony Ng
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Carol D Jones
- Department of Pathology, Stanford University, Stanford, California, USA
| | - Sushama Varma
- Department of Pathology, Stanford University, Stanford, California, USA
| | - Justin I Odegaard
- Department of Pathology, Stanford University, Stanford, California, USA
| | - Toshihiro Sugiyama
- Department of Biochemistry, Akita University Graduate School of Medicine, Akita, Japan
| | - Souichi Koyota
- Department of Biochemistry, Akita University Graduate School of Medicine, Akita, Japan
| | - Brian P Rubin
- Department of Anatomic Pathology, Cleveland Clinic, Cleveland, Ohio, USA
| | - Megan L Troxell
- Department of Pathology, Oregon Health and Sciences University, Portland, Oregon, USA
| | | | - James L Zehnder
- Department of Pathology, Stanford University, Stanford, California, USA
| | - Philip A Beachy
- 1] Department of Biochemistry, Stanford University, Stanford, California, USA. [2] Department of Developmental Biology, Stanford University, Stanford, California, USA. [3] Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, California, USA. [4] Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, California, USA
| | | | - Robert B West
- Department of Pathology, Stanford University, Stanford, California, USA
| |
Collapse
|
121
|
Genetic targeting of B-RafV600E affects survival and proliferation and identifies selective agents against BRAF-mutant colorectal cancer cells. Mol Cancer 2014; 13:122. [PMID: 24885690 PMCID: PMC4035728 DOI: 10.1186/1476-4598-13-122] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2013] [Accepted: 05/09/2014] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Colorectal cancers carrying the B-Raf V600E-mutation are associated with a poor prognosis. The purpose of this study was to identify B-RafV600E-mediated traits of cancer cells in a genetic in vitro model and to assess the selective sensitization of B-RafV600E-mutant cancer cells towards therapeutic agents. METHODS Somatic cell gene targeting was used to generate subclones of the colorectal cancer cell line RKO containing either wild-type or V600E-mutant B-Raf kinase. Cell-biologic analyses were performed in order to link cancer cell traits to the BRAF-mutant genotype. Subsequently, the corresponding tumor cell clones were characterized pharmacogenetically to identify therapeutic agents exhibiting selective sensitivity in B-RafV600E-mutant cells. RESULTS Genetic targeting of mutant BRAF resulted in restoration of sensitivity to serum starvation-induced apoptosis and efficiently inhibited cell proliferation in the absence of growth factors. Among tested agents, the B-Raf inhibitor dabrafenib was found to induce a strong V600E-dependent shift in cell viability. In contrast, no differential sensitizing effect was observed for conventional chemotherapeutic agents (mitomycin C, oxaliplatin, paclitaxel, etoposide, 5-fluorouracil), nor for the targeted agents cetuximab, sorafenib, vemurafenib, RAF265, or for inhibition of PI3 kinase. Treatment with dabrafenib efficiently inhibited phosphorylation of the B-Raf downstream targets Mek 1/2 and Erk 1/2. CONCLUSION Mutant BRAF alleles mediate self-sufficiency of growth signals and serum starvation-induced resistance to apoptosis. Targeting of the BRAF mutation leads to a loss of these hallmarks of cancer. Dabrafenib selectively inhibits cell viability in B-RafV600E mutant cancer cells.
Collapse
|
122
|
Leto SM, Trusolino L. Primary and acquired resistance to EGFR-targeted therapies in colorectal cancer: impact on future treatment strategies. J Mol Med (Berl) 2014; 92:709-22. [PMID: 24811491 PMCID: PMC4055851 DOI: 10.1007/s00109-014-1161-2] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2014] [Revised: 04/28/2014] [Accepted: 04/29/2014] [Indexed: 12/23/2022]
Abstract
Only approximately 10 % of genetically unselected patients with chemorefractory metastatic colorectal cancer experience tumor regression when treated with the anti-epidermal growth factor receptor (EGFR) antibodies cetuximab or panitumumab (“primary” or “de novo” resistance). Moreover, nearly all patients whose tumors initially respond inevitably become refractory (“secondary” or “acquired” resistance). An ever-increasing number of predictors of both primary and acquired resistance to anti-EGFR antibodies have been described, and it is now evident that most of the underlying mechanisms significantly overlap. By trying to extrapolate a unifying perspective out of many idiosyncratic details, here, we discuss the molecular underpinnings of therapeutic resistance, summarize research efforts aimed to improve patient selection, and present alternative therapeutic strategies that are now under development to increase response and combat relapse.
Collapse
Affiliation(s)
- Simonetta M Leto
- Department of Oncology, University of Torino Medical School, 10060, Candiolo, Torino, Italy
| | | |
Collapse
|
123
|
Capalbo C, Marchetti P, Coppa A, Calogero A, Anastasi E, Buffone A, Belardinilli F, Gulino M, Frati P, Catalano C, Cortesi E, Giannini G, Gulino A. Vemurafenib and panitumumab combination tailored therapy in BRAF-mutated metastatic colorectal cancer: a case report. Cancer Biol Ther 2014; 15:826-31. [PMID: 24755613 PMCID: PMC4100983 DOI: 10.4161/cbt.28878] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
As the knowledge on cancer genetic alterations progresses, it fosters the need for more personalized therapeutic intervention in modern cancer management. Recently, mutations in KRAS, BRAF, and PIK3CA genes have emerged as important mechanisms of resistance to EGFR-targeted therapy in metastatic colorectal cancer (mCRC). Here we report the first case of a mCRC patient whose disease had progressed on standard lines of treatment and for which we devised a personalized therapeutic approach consisting of vemurafenib (Zelboraf) and panitumumab (Vectibix), based on the following molecular profile: BRAF(V600E)-mutant, amplified EGFR (double positive) and WT KRAS, WT PIK3CA, not-amplified HER2 (triple negative). This new combination therapy was well tolerated and resulted in a strong control of the disease. In particular, the vemurafenib-panitumumab combination appears to limit the typical toxicity of single agents, since no cutaneous toxic effects typically associated with vemurafenib were observed. Here we report the first clinical evidence that the combination of an anti-EGFR (panitumumab) and an inhibitor of BRAF(V600E) (vemurafenib) is well tolerated and results in a strong disease control in an extensively pretreated mCRC patient.
Collapse
Affiliation(s)
- Carlo Capalbo
- Department of Molecular Medicine; Sapienza University of Rome; Rome, Italy; Medical Oncology; IDI IRCCS; Rome, Italy
| | - Paolo Marchetti
- Medical Oncology; IDI IRCCS; Rome, Italy; Department of Clinical and Molecular Medicine; Sapienza University of Rome; Rome, Italy
| | - Anna Coppa
- Department of Experimental Medicine; Sapienza University of Rome; Rome, Italy
| | - Antonella Calogero
- Department of Medical-Surgical Sciences and Biotechnologies; Sapienza University of Rome; Rome, Italy
| | - Emanuela Anastasi
- Department of Molecular Medicine; Sapienza University of Rome; Rome, Italy
| | - Amelia Buffone
- Department of Molecular Medicine; Sapienza University of Rome; Rome, Italy
| | | | - Matteo Gulino
- Department of Anatomical, Histological, Forensic Medicine and Orthopedic Science; Forensic Medicine Section; Sapienza University of Rome; Rome, Italy
| | - Paola Frati
- Department of Anatomical, Histological, Forensic Medicine and Orthopedic Science; Forensic Medicine Section; Sapienza University of Rome; Rome, Italy; Neuromed IRCCS; Pozzilli, Italy
| | - Carlo Catalano
- Department of Radiological, Oncological and Pathological Sciences; Sapienza University of Rome; Rome, Italy
| | - Enrico Cortesi
- Department of Radiological, Oncological and Pathological Sciences; Sapienza University of Rome; Rome, Italy
| | - Giuseppe Giannini
- Department of Molecular Medicine; Sapienza University of Rome; Rome, Italy
| | - Alberto Gulino
- Neuromed IRCCS; Pozzilli, Italy; Department of Molecular Medicine; Center for Life Nanoscience@Sapienza; Italian Institute of Technology and Istituto Pasteur; Fondazione Cenci-Bolognetti; Sapienza University of Rome; Rome, Italy
| |
Collapse
|
124
|
Farrell CL, Pedigo NG, Messersmith AR. Application of genomic principles to pharmacotherapy of cancer. AMERICAN JOURNAL OF PHARMACEUTICAL EDUCATION 2014; 78:55. [PMID: 24761016 PMCID: PMC3996387 DOI: 10.5688/ajpe78355] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2013] [Accepted: 10/08/2013] [Indexed: 05/09/2023]
Abstract
OBJECTIVES To teach first-year (P1) pharmacy students to apply the principles of pharmacogenomics underlying clinical pharmacotherapeutics to cancer patients. DESIGN Using polymerase chain reaction (PCR) and high-resolution melting analysis of deoxyribonucleic acid (DNA) from colorectal cancer cell lines to determine the presence of somatic mutations for an oncogenic marker, students formulated the proper course of treatment for a patient with similar tumor genomics. ASSESSMENT In a postintervention survey, students highly rated the effectiveness of the laboratory session for learning pharmacogenomics, and subsequent examination scores reflected retention of principles and understanding of clinical application. CONCLUSION The pharmacogenomic laboratory exercise prepared students to understand how genetic markers give clinical insight into the appropriate application of drugs in oncology pharmacotherapy. Further, the session inspired their interest in learning more about pharmacogenomics and their professional roles in personalized medicine.
Collapse
Affiliation(s)
| | - Nancy G Pedigo
- School of Pharmacy, Presbyterian College, Clinton, South Carolina
| | | |
Collapse
|
125
|
Goffin JR, Zbuk K. Epidermal growth factor receptor: pathway, therapies, and pipeline. Clin Ther 2014; 35:1282-303. [PMID: 24054705 DOI: 10.1016/j.clinthera.2013.08.007] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2013] [Revised: 08/16/2013] [Accepted: 08/21/2013] [Indexed: 12/15/2022]
Abstract
BACKGROUND The epidermal growth factor receptor (EGFR) pathway is important in tumor growth, survival, and metastasis and is now the target of several therapeutic agents. OBJECTIVES This paper seeks to review the EGFR pathway, the study and use of EGFR-directed agents in non-small-cell lung cancer (NSCLC) and colorectal cancer (CRC), and related new drug development. METHODS PubMed was searched for English-language articles by MeSH and title terms of EGFR published from 2006 to 2013, using the limits of clinical trials as well as reviews. Reference lists were assessed for relevant articles, and guidelines were searched. Clinicaltrials.gov and meeting abstracts were queried for investigational agents. Eligible papers included those concerning EGFR biology, NSCLC or CRC studies involving EGFR-directed agents, and/or investigational drugs targeting EGFR and/or associated pathways. RESULTS The activity of oral tyrosine kinase inhibitors (TKIs) against EGFR has improved survival in NSCLC, and these agents particularly effective in cancers with an EGFR mutation. Resistance to TKIs is most commonly related to a second, T790M, mutation, or to MET amplification, with newer agents directed against these mechanisms. Conversely, in CRC, TKIs have been ineffective, whereas monoclonal antibodies have improved survival. Both primary and secondary KRAS mutations in CRC abrogate mAb effectiveness. Several targets, including MET, BRAF, and PI3K, may serve useful in combination with anti-EGFR drugs. CONCLUSIONS Exploitation of EGFR-directed therapies has offered improvement in survival and quality of life in NSCLC and CRC. New therapies directed at EGFR may offer further improvements. However, resistance mechanisms suggest that combination therapies or multitargeted agents will be crucial in making significant future advances.
Collapse
Affiliation(s)
- John R Goffin
- Department of Oncology, Faculty of Health Sciences, McMaster University, Hamilton, Canada.
| | | |
Collapse
|
126
|
Abstract
In multiple myeloma, it is believed that multiple mutations in different pathways deregulate the intrinsic biology of the plasma cell, resulting in a genetically complex heterogeneous disease. Mutations in the mitogen-activated protein kinase/extracellular signal-regulated kinase pathway have been identified and represent potential targets for therapy in this disease. BRAF, a serine/threonine kinase, has received considerable attention given the success of targeted therapy in malignant melanoma. Andrulis and colleagues report, for the first time, successful treatment of multiple myeloma with vemurafenib, a BRAF inhibitor, in a patient with a BRAF mutation.
Collapse
Affiliation(s)
- Elizabeth O'Donnell
- Massachusetts General Hospital Cancer Center, Massachusetts General Hospital, Boston, MA 02114, USA
| | | |
Collapse
|
127
|
Connolly K, Brungs D, Szeto E, Epstein RJ. Anticancer activity of combination targeted therapy using cetuximab plus vemurafenib for refractory BRAF (V600E)-mutant metastatic colorectal carcinoma. ACTA ACUST UNITED AC 2014; 21:e151-4. [PMID: 24523613 DOI: 10.3747/co.21.1661] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Mismatch-repair-deficient colorectal cancers often contain kinase-activating V600E BRAF mutations, but no clinical utility has yet been demonstrated in this setting for monotherapy using oral braf kinase inhibitors such as vemurafenib or dabrafenib. Recent studies have indicated that tumour resistance to braf inhibition is mediated by upregulated epidermal growth factor receptor (egfr) signalling, disruption of which is a routine treatment strategy in KRAS wild-type colorectal cancer. In this report, we describe the clinical course of a heavily pretreated patient who elected to receive off-label dual-targeted braf- and egfr-inhibitory therapy with good tolerance and apparent clinical benefit.
Collapse
Affiliation(s)
- K Connolly
- Department of Oncology, The Kinghorn Cancer Centre, St Vincent's Hospital, and UNSW Clinical School, Sydney, Australia
| | - D Brungs
- Department of Oncology, The Kinghorn Cancer Centre, St Vincent's Hospital, and UNSW Clinical School, Sydney, Australia
| | - E Szeto
- Department of Oncology, The Kinghorn Cancer Centre, St Vincent's Hospital, and UNSW Clinical School, Sydney, Australia
| | - R J Epstein
- Department of Oncology, The Kinghorn Cancer Centre, St Vincent's Hospital, and UNSW Clinical School, Sydney, Australia
| |
Collapse
|
128
|
Abstract
BRAF represents one of the most frequently mutated protein kinase genes in human tumours. The mutation is commonly tested in pathology practice. BRAF mutation is seen in melanoma, papillary thyroid carcinoma (including papillary thyroid carcinoma arising from ovarian teratoma), ovarian serous tumours, colorectal carcinoma, gliomas, hepatobiliary carcinomas and hairy cell leukaemia. In these cancers, various genetic aberrations of the BRAF proto-oncogene, such as different point mutations and chromosomal rearrangements, have been reported. The most common mutation, BRAF V600E, can be detected by DNA sequencing and immunohistochemistry on formalin fixed, paraffin embedded tumour tissue. Detection of BRAF V600E mutation has the potential for clinical use as a diagnostic and prognostic marker. In addition, a great deal of research effort has been spent in strategies inhibiting its activity. Indeed, recent clinical trials involving BRAF selective inhibitors exhibited promising response rates in metastatic melanoma patients. Clinical trials are underway for other cancers. However, cutaneous side effects of treatment have been reported and therapeutic response to cancer is short-lived due to the emergence of several resistance mechanisms. In this review, we give an update on the clinical pathological relevance of BRAF mutation in cancer. It is hoped that the review will enhance the direction of future research and assist in more effective use of the knowledge of BRAF mutation in clinical practice.
Collapse
|
129
|
Abstract
Activating BRAF mutations, leading to constitutive activation of the MAPK signaling pathway, are common in a variety of human cancers. Several small molecule BRAF inhibitors have been developed during the last years and shown promising results in clinical trials, especially for metastatic melanoma, while they have been less effective in colon cancer. Two inhibitors, vemurafenib and dabrafenib, have been approved for treatment of melanoma. Unfortunately, in most patients who initially respond the tumors eventually develop acquired resistance to the BRAF inhibitors. So far, a number of resistance mechanisms have been identified, including secondary NRAS mutations and BRAF alternative splicing, leading to reactivation of the MAPK pathway. Other alterations, both upstream and downstream of BRAF can have the same effect, and activation of alternative pathways can also play a role in resistance to BRAF inhibitors. In addition, intra-tumor heterogeneity with the presence of clones of tumor cells lacking BRAF mutations needs to be considered, since wildtype BRAF can be activated by inhibitors designed to target mutated BRAF. Combination of the BRAF inhibitor dabrafenib with the MEK inhibitor trametinib has significantly prolonged progression free survival compared to dabrafenib alone in metastatic melanoma. Combination treatments of BRAF inhibitors with other agents may not only circumvent or delay resistance, but may also lead to fewer side effects, such as development of secondary squamous tumors. Several clinical trials are underway for many different BRAF mutation positive cancers with BRAF inhibitors alone or in combination with other small molecule inhibitors, immunotherapies or conventional chemotherapy.
Collapse
|
130
|
Clancy C, Burke JP, Kalady MF, Coffey JC. BRAF mutation is associated with distinct clinicopathological characteristics in colorectal cancer: a systematic review and meta-analysis. Colorectal Dis 2013; 15:e711-8. [PMID: 24112392 DOI: 10.1111/codi.12427] [Citation(s) in RCA: 88] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2013] [Accepted: 06/18/2013] [Indexed: 12/22/2022]
Abstract
AIM Colorectal cancer is a heterogeneous disease with multiple underlying genetic mutations resulting in different phenotypes. Mutation in the v-Raf murine sarcoma viral oncogene homolog B1 (BRAF) proto-oncogene is an important event in the methylator pathway. There is no consensus, however, on the clinicopathological characteristics associated with BRAF mutation. METHOD A comprehensive search for published studies examining the effect of BRAF mutation on colorectal cancer was performed. Random effects methods were used to combine data. RESULTS Data were retrieved from 21 studies describing 9885 patients. BRAF associated colorectal cancer is associated with proximal tumour location (OR 5.222, 95% CI 3.801-7.174, P < 0.001), T4 tumours (OR 1.761, 95% CI 1.164-2.663, P = 0.007) and poor differentiation (OR 3.816, 95% CI 2.714-5.365, P < 0.001) and is negatively associated with male sex (OR 0.623, 95% CI 0.505-0.769, P < 0.001), age of diagnosis under 60 years (OR 0.453, 95% CI 0.280-0.733, P = 0.001) and rectal cancer (OR 0.266, 95% CI 0.122-0.422, P < 0.001). CONCLUSION BRAF mutation appears to be associated with distinct, unfavourable clinicopathological characteristics in colorectal cancer.
Collapse
Affiliation(s)
- C Clancy
- Department of Colorectal Surgery, University Hospital Limerick, Limerick, Ireland
| | | | | | | |
Collapse
|
131
|
Kim SH, Kang JG, Kim CS, Ihm SH, Choi MG, Yoo HJ, Lee SJ. Akt inhibition enhances the cytotoxic effect of apigenin in combination with PLX4032 in anaplastic thyroid carcinoma cells harboring BRAFV600E. J Endocrinol Invest 2013; 36:1099-104. [PMID: 24084189 DOI: 10.3275/9099] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Aim of the present study was to evaluate the effect of apigenin in combination with BRAFV600E inhibitor PLX4032 on cell survival, and to investigate the influence of Akt inhibition on the combined effect of apigenin and PLX4032 in ATC cells harboring BRAFV600E. In 8505C and FRO cells harboring BRAFV600E, after treatment of apigenin and PLX4032, the cell viability decreased, and the percentage of dead cells increased in a time- and concentration-dependent manner, respectively. In apigenin- and PLX4032- treated cells, compared with apigenin alone-treated cells, the cell viability was lessened, and the percentage of dead cells was multiplied. In the addition of PLX4032 to apigenin, compared with the treatment of apigenin alone, the protein levels of cleaved PARP-1 and cleaved caspase-3 were elevated, and phospho-ERK protein levels were reduced, and the protein levels of total ERK, c-Myc, BRAF, phospho-Akt, phospho-p70S6K and phospho-4EBP1 were not varied. Compared with the treatment of PLX4032 alone, phosphop70S6K protein levels were reduced, and the other protein levels were not altered. Phospho-ERK protein levels were reduced only in 8505C cells. Under the co-treatment of apigenin and PLX4032, administration of the PI3K inhibitor wortmannin further decreased the cell viability, and increased the percentage of dead cells. In conclusion, our results suggest that PLX4032 augments apigenin-induced cytotoxicity in ATC cells harboring BRAFV600E. Moreover, Akt suppression potentiates the combined effect of apigenin and PLX4032 in ATC cells harboring BRAFV600E.
Collapse
Affiliation(s)
- S H Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, Hallym University, Chuncheon 200-704, Republic of Korea
| | | | | | | | | | | | | |
Collapse
|
132
|
Peng W, Hu J, Zhu XD, Liu X, Wang CC, Li WH, Chen ZY. Overexpression of miR-145 increases the sensitivity of vemurafenib in drug-resistant colo205 cell line. Tumour Biol 2013; 35:2983-8. [PMID: 24248543 DOI: 10.1007/s13277-013-1383-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2013] [Accepted: 11/05/2013] [Indexed: 12/18/2022] Open
Abstract
Vemurafenib is a selective and potent small molecule inhibitor of the V600 mutant form of the BRAF protein used in the treatment of melanoma and colorectal cancer. However, vemurafenib has less effect in BRAF mutant colorectal cancer due to the resistance of tumor cell to vemurafenib. To verify whether or not miR-145, a short RNA molecule of microRNA which has been supposed to be a tumor suppressor, is involved in this process, we established vemurafenib-resistant cell line colo205/V and found that the miR-145 expression was significantly downregulated in colo205/V cells compared to normal colo205 cells. Moreover, the overexpression of miR-145 could increase the sensitivity of colo205/V cells to vemurafenib both in vitro and in vivo. In conclusion, miR-145 might be used as a therapeutic target in the treatment of colorectal cancer patients with BRAF V600E mutation.
Collapse
Affiliation(s)
- Wei Peng
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, 270 Dong An Road, Shanghai, 200032, China
| | | | | | | | | | | | | |
Collapse
|
133
|
Wang J, Chen J, Miller DD, Li W. Synergistic combination of novel tubulin inhibitor ABI-274 and vemurafenib overcome vemurafenib acquired resistance in BRAFV600E melanoma. Mol Cancer Ther 2013; 13:16-26. [PMID: 24249714 DOI: 10.1158/1535-7163.mct-13-0212] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Acquired clinical resistance to vemurafenib, a selective BRAF(V600E) inhibitor, arises frequently after short-term chemotherapy. Because inhibitions of targets in the RAF-MEK-ERK pathway result in G(0)-G(1) cell-cycle arrest, vemurafenib-resistant cancer cells are expected to escape this cell-cycle arrest and progress to the subsequent G(2)-M phase. We hypothesized that a combined therapy using vemurafenib with a G(2)-M phase blocking agent will trap resistant cells and overcome vemurafenib resistance. To test this hypothesis, we first determined the combination index (CI) values of our novel tubulin inhibitor ABI-274 and vemurafenib on parental human A375 and MDA-MB-435 melanoma cell lines to be 0.32 and 0.1, respectively, suggesting strong synergy for the combination. We then developed an A375RF21 subline with significant acquired resistance to vemurafenib and confirmed the strong synergistic effect. Next, we studied the potential mechanisms of overcoming vemurafenib resistance. Flow cytometry confirmed that the combination of ABI-274 and vemurafenib synergistically arrested cells in the G(1)-G(2)-M phase, and significantly increased apoptosis in both parental A375 and the vemurafenib-resistant A375RF21 cells. Western blot analysis revealed that the combination treatment effectively reduced the level of phosphorylated and total AKT, activated the apoptosis cascade, and increased cleaved caspase-3 and cleaved PARP, but had no significant influence on the level of extracellular signal-regulated kinase (ERK) phosphorylation. Finally, in vivo coadministration of vemurafenib with ABI-274 showed strong synergistic efficacy in the vemurafenib-resistant xenograft model in nude mice. Overall, these results offer a rational combination strategy to significantly enhance the therapeutic benefit in patients with melanoma who inevitably become resistant to current vemurafenib therapy.
Collapse
Affiliation(s)
- Jin Wang
- Corresponding Author: Wei Li, The University of Tennessee Health Science Center, Room 327B, 847 Monroe Ave, Memphis, TN 38163.
| | | | | | | |
Collapse
|
134
|
Thiel A, Ristimäki A. Toward a Molecular Classification of Colorectal Cancer: The Role of BRAF. Front Oncol 2013; 3:281. [PMID: 24298448 PMCID: PMC3828559 DOI: 10.3389/fonc.2013.00281] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2013] [Accepted: 10/31/2013] [Indexed: 12/24/2022] Open
Abstract
Different genetic aberrations of BRAF have been reported in various malignancies. BRAF is member of the RAS/RAF/MEK/ERK pathway and constitutive activity of this pathway can lead to increased cellular growth, invasion, and metastasis. The most common activating BRAF mutation in colorectal cancer is the V600E mutation, which is present in 5–15% of all tumors, and up to 80% of tumors with high microsatellite instability (MSI) harbor this mutation. BRAF mutation is associated with proximal location, higher age, female gender, MSI-H, high grade, and mucinous histology, and is a marker of poor prognosis in colorectal cancer. The role of BRAF mutation as a predictive marker in respect of EGFR targeted treatments is controversial. BRAF V600 selective inhibitors have been approved for the treatment of V600 mutation positive metastatic melanoma, but the response rates in colorectal cancer are poor. This might be due to innate resistance mechanisms of colorectal cancers against the treatment solely targeting BRAF. To overcome resistance the combination of treatments, simultaneous inhibition of BRAF and MEK or PI3K/mTOR, might emerge as a successful therapeutic concept.
Collapse
Affiliation(s)
- Alexandra Thiel
- Division of Pathology, HUSLAB and Haartman Institute, Helsinki University Central Hospital, University of Helsinki , Helsinki , Finland ; Genome-Scale Biology, Research Programs Unit, University of Helsinki , Helsinki , Finland
| | | |
Collapse
|
135
|
Lito P, Rosen N, Solit DB. Tumor adaptation and resistance to RAF inhibitors. Nat Med 2013; 19:1401-9. [DOI: 10.1038/nm.3392] [Citation(s) in RCA: 446] [Impact Index Per Article: 37.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2013] [Accepted: 10/07/2013] [Indexed: 12/12/2022]
|
136
|
Kin C, Kidess E, Poultsides GA, Visser BC, Jeffrey SS. Colorectal cancer diagnostics: biomarkers, cell-free DNA, circulating tumor cells and defining heterogeneous populations by single-cell analysis. Expert Rev Mol Diagn 2013; 13:581-99. [PMID: 23895128 DOI: 10.1586/14737159.2013.811896] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Reliable biomarkers are needed to guide treatment of colorectal cancer, as well as for surveillance to detect recurrence and monitor therapeutic response. In this review, the authors discuss the use of various biomarkers in addition to serum carcinoembryonic antigen, the current surveillance method for metastatic recurrence after resection. The clinical relevance of mutations including microsatellite instability, KRAS, BRAF and SMAD4 is addressed. The role of circulating tumor cells and cell-free DNA with regards to their implementation into clinical use is discussed, as well as how single-cell analysis may fit into a monitoring program. The detection and characterization of circulating tumor cells and cell-free DNA in colorectal cancer patients will not only improve the understanding of the development of metastasis, but may also supplant the use of other biomarkers.
Collapse
Affiliation(s)
- Cindy Kin
- Department of Surgery, Stanford University School of Medicine, CA, USA.
| | | | | | | | | |
Collapse
|
137
|
Loupakis F, Cremolini C, Salvatore L, Masi G, Sensi E, Schirripa M, Michelucci A, Pfanner E, Brunetti I, Lupi C, Antoniotti C, Bergamo F, Lonardi S, Zagonel V, Simi P, Fontanini G, Falcone A. FOLFOXIRI plus bevacizumab as first-line treatment in BRAF mutant metastatic colorectal cancer. Eur J Cancer 2013; 50:57-63. [PMID: 24138831 DOI: 10.1016/j.ejca.2013.08.024] [Citation(s) in RCA: 137] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2013] [Revised: 08/27/2013] [Accepted: 08/28/2013] [Indexed: 01/02/2023]
Abstract
BACKGROUND BRAF V600E mutation plays a negative prognostic role in metastatic colorectal cancer (mCRC), leading to a median Progression Free Survival (PFS) of 4-6months with first-line conventional treatments. Our group recently reported in a retrospective exploratory analysis of a phase II trial that FOLFOXIRI (5-FU/LV+Oxaliplatin+Irinotecan) plus bevacizumab might allow to achieve remarkable results in terms of PFS and Overall Survival (OS) also in this poor-prognosis subgroup. The aim of this work was to prospectively validate our retrospective finding. PATIENTS AND METHODS This phase II trial was designed to detect an increase in 6month-Progression Free Rate (6m-PFR) from 45% to 80% in a population of BRAF mutant mCRC patients treated with first-line FOLFOXIRI plus bevacizumab. Secondary end-points were PFS, OS, response rate (RR) and the analysis of outcome parameters in the pooled population consisting of both retrospectively and prospectively included patients. This trial is registered with ClinicalTrials.gov, number NCT01437618. RESULTS Two-hundred-fourteen potentially eligible mCRC patients were screened for BRAF mutational status. Fifteen BRAF mutant patients (7%) were included in the validation cohort. At a median follow up of 25.7months, 6m-PFR was 73%. Median PFS and OS were 9.2 and 24.1months, respectively. In the pooled population, at a median follow up of 40.4months, 6m-PFR was 84%. Median PFS and OS were 11.8 and 24.1months, respectively. Overall RR and disease control rate were 72% and 88%, respectively. CONCLUSION Lacking randomised trials in this specific molecular subgroup, FOLFOXIRI plus bevacizumab might be a reasonable option for the first-line treatment of BRAF mutant mCRC patients.
Collapse
Affiliation(s)
- F Loupakis
- Unit of Medical Oncology 2, Azienda Ospedaliera-Universitaria Pisana, Via Roma 67, 56126 Pisa, Italy.
| | - C Cremolini
- Unit of Medical Oncology 2, Azienda Ospedaliera-Universitaria Pisana, Via Roma 67, 56126 Pisa, Italy
| | - L Salvatore
- Unit of Medical Oncology 2, Azienda Ospedaliera-Universitaria Pisana, Via Roma 67, 56126 Pisa, Italy
| | - G Masi
- Unit of Medical Oncology 2, Azienda Ospedaliera-Universitaria Pisana, Via Roma 67, 56126 Pisa, Italy
| | - E Sensi
- Division of Pathology, Department of Surgery, University of Pisa, Via Roma 67, 56126 Pisa, Italy
| | - M Schirripa
- Unit of Medical Oncology 2, Azienda Ospedaliera-Universitaria Pisana, Via Roma 67, 56126 Pisa, Italy
| | - A Michelucci
- Unit of Medical Genetics, Azienda Ospedaliera-Universitaria Pisana, Via Roma 67, 56126 Pisa, Italy
| | - E Pfanner
- Unit of Medical Oncology 2, Azienda Ospedaliera-Universitaria Pisana, Via Roma 67, 56126 Pisa, Italy
| | - I Brunetti
- Unit of Medical Oncology 1, Azienda Ospedaliera-Universitaria Pisana, Via Roma 67, 56126 Pisa, Italy
| | - C Lupi
- Division of Pathology, Department of Surgery, University of Pisa, Via Roma 67, 56126 Pisa, Italy
| | - C Antoniotti
- Unit of Medical Oncology 2, Azienda Ospedaliera-Universitaria Pisana, Via Roma 67, 56126 Pisa, Italy
| | - F Bergamo
- Unit of Medical Oncology 1, Oncology Institute of Veneto, Via Gattamelata 64, 35138 Padua, Italy
| | - S Lonardi
- Unit of Medical Oncology 1, Oncology Institute of Veneto, Via Gattamelata 64, 35138 Padua, Italy
| | - V Zagonel
- Unit of Medical Oncology 1, Oncology Institute of Veneto, Via Gattamelata 64, 35138 Padua, Italy
| | - P Simi
- Unit of Medical Genetics, Azienda Ospedaliera-Universitaria Pisana, Via Roma 67, 56126 Pisa, Italy
| | - G Fontanini
- Division of Pathology, Department of Surgery, University of Pisa, Via Roma 67, 56126 Pisa, Italy
| | - A Falcone
- Unit of Medical Oncology 2, Azienda Ospedaliera-Universitaria Pisana, Via Roma 67, 56126 Pisa, Italy; Division of Medical Oncology, Department of Oncology, Transplants and New Technologies in Medicine, University of Pisa, Via Roma 67, 56126 Pisa, Italy
| |
Collapse
|
138
|
Petanidis S, Anestakis D, Argyraki M, Hadzopoulou-Cladaras M, Salifoglou A. Differential expression of IL-17, 22 and 23 in the progression of colorectal cancer in patients with K-ras mutation: Ras signal inhibition and crosstalk with GM-CSF and IFN-γ. PLoS One 2013; 8:e73616. [PMID: 24040001 PMCID: PMC3765247 DOI: 10.1371/journal.pone.0073616] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2013] [Accepted: 07/23/2013] [Indexed: 12/13/2022] Open
Abstract
Recent studies have suggested that aberrant K-ras signaling is responsible for triggering immunological responses and inflammation-driven tumorigenesis. Interleukins IL-17, IL-22, and IL-23 have been reported in various types of malignancies, but the exact mechanistic role of these molecules remains to be elucidated. Given the role of K-ras and the involvement of interleukins in colorectal tumorigenesis, research efforts are reported for the first time, showing that differentially expressed interleukin IL-17, IL-22, and IL-23 levels are associated with K-ras in a stage-specific fashion along colorectal cancer progression. Specifically, a) the effect of K-ras signaling was investigated in the overall expression of interleukins in patients with colorectal cancer and healthy controls, and b) an association was established between mutant K-ras and cytokines GM-CSF and IFN-γ. The results indicate that specific interleukins are differentially expressed in K-ras positive patients and the use of K-ras inhibitor Manumycin A decreases both interleukin levels and apoptosis in Caco-2 cells by inhibiting cell viability. Finally, inflammation-driven GM-CSF and IFN-γ levels are modulated through interleukin expression in tumor patients, with interleukin expression in the intestinal lumen and cancerous tissue mediated by aberrant K-ras signaling. Collectively, the findings a) indicate that interleukin expression is influenced by ras signaling and specific interleukins play an oncogenic promoter role in colorectal cancer, highlighting the molecular link between inflammation and tumorigenesis, and b) accentuate the interwoven molecular correlations as leads to new therapeutic approaches in the future.
Collapse
Affiliation(s)
- Savvas Petanidis
- Department of Chemical Engineering, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Doxakis Anestakis
- Laboratory of General Biology, Medical School, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Maria Argyraki
- Laboratory of General Biology, Medical School, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Margarita Hadzopoulou-Cladaras
- Department of Genetics, Development and Molecular Biology, School of Biology, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Athanasios Salifoglou
- Department of Chemical Engineering, Aristotle University of Thessaloniki, Thessaloniki, Greece
| |
Collapse
|
139
|
Landau DA, Carter SL, Getz G, Wu CJ. Clonal evolution in hematological malignancies and therapeutic implications. Leukemia 2013; 28:34-43. [PMID: 23979521 DOI: 10.1038/leu.2013.248] [Citation(s) in RCA: 122] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2013] [Revised: 07/22/2013] [Accepted: 08/14/2013] [Indexed: 12/19/2022]
Abstract
The ability of cancer to evolve and adapt is a principal challenge to therapy in general and to the paradigm of targeted therapy in particular. This ability is fueled by the co-existence of multiple, genetically heterogeneous subpopulations within the cancer cell population. Increasing evidence has supported the idea that these subpopulations are selected in a Darwinian fashion, by which the genetic landscape of the tumor is continuously reshaped. Massively parallel sequencing has enabled a recent surge in our ability to study this process, adding to previous efforts using cytogenetic methods and targeted sequencing. Altogether, these studies reveal the complex evolutionary trajectories occurring across individual hematological malignancies. They also suggest that while clonal evolution may contribute to resistance to therapy, treatment may also hasten the evolutionary process. New insights into this process challenge us to understand the impact of treatment on clonal evolution and inspire the development of novel prognostic and therapeutic strategies.
Collapse
Affiliation(s)
- D A Landau
- 1] Cancer Vaccine Center, Dana-Farber Cancer Institute, Boston, MA, USA [2] Broad Institute, Cambridge, MA, USA [3] Department of Hematology, Yale Cancer Center, New Haven, CT, USA [4] Université Paris Diderot, Paris, France
| | | | - G Getz
- 1] Broad Institute, Cambridge, MA, USA [2] Massachusetts General Hospital Cancer Center and Department of Pathology, Boston, MA, USA
| | - C J Wu
- 1] Cancer Vaccine Center, Dana-Farber Cancer Institute, Boston, MA, USA [2] Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA [3] Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
140
|
Kline CLB, El-Deiry WS. Personalizing colon cancer therapeutics: targeting old and new mechanisms of action. Pharmaceuticals (Basel) 2013; 6:988-1038. [PMID: 24276379 PMCID: PMC3817731 DOI: 10.3390/ph6080988] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2013] [Revised: 07/30/2013] [Accepted: 08/16/2013] [Indexed: 12/17/2022] Open
Abstract
The use of pharmaceuticals for colon cancer treatment has been increasingly personalized, in part due to the development of new molecular tools. In this review, we discuss the old and new colon cancer chemotherapeutics, and the parameters that have been shown to be predictive of efficacy and safety of these chemotherapeutics. In addition, we discuss how alternate pharmaceuticals have been developed in light of a potential lack of response or resistance to a particular chemotherapeutic.
Collapse
Affiliation(s)
- Christina Leah B Kline
- Hematology/Oncology Division, Penn State Hershey Medical Center, Hershey, PA 17033, USA.
| | | |
Collapse
|
141
|
Minuti G, D'Incecco A, Cappuzzo F. Targeted therapy for NSCLC with driver mutations. Expert Opin Biol Ther 2013; 13:1401-12. [PMID: 23930754 DOI: 10.1517/14712598.2013.827657] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
INTRODUCTION Activating mutations of the epidermal growth factor receptor (EGFR) gene and rearrangement of anaplastic lymphoma kinase (ALK) gene best illustrate the therapeutic relevance of molecular characterization in non-small cell lung cancer (NSCLC) patients. Several genetic aberrations with a potential prognostic or predictive role have been identified, mainly in adenocarcinoma subtype, including ROS1, RET, MET, HER2, BRAF and KRAS. More recently oncogenic drivers, such as DDR2, FGFR1 and PI3KCA, have been characterized in squamous cell lung carcinoma (SCC) and target agents are currently under evaluation. The aim of this review is to summarize the growing scenario of new targetable oncogenes in NSCLC. AREAS COVERED For this review article all published data on NSCLC genomic alterations, including the techniques employed for oncogenic drivers identification, the prevalence of each one in lung cancer subtypes, the preclinical data corroborating their role in tumorigenesis and the potential biological tailored agents tested and under evaluation were collected and analyzed using PubMed. EXPERT OPINION Oncogenic products represent reliable targets for drug therapy and the expanding knowledge of molecular pathways involved in lung tumorigenesis is resulting in a dramatic change of treatment strategies leading to an improvement in disease and symptom control, extending life duration and improving quality of life.
Collapse
Affiliation(s)
- Gabriele Minuti
- Medical Oncology Department, Civil Hospital of Livorno, Istituto Toscano Tumori , Viale Alfieri 36, 57100, Livorno , Italy +39 0586 223189 ; +39 0586 223457 ;
| | | | | |
Collapse
|
142
|
Chen J, Shao R, Zhang XD, Chen C. Applications of nanotechnology for melanoma treatment, diagnosis, and theranostics. Int J Nanomedicine 2013; 8:2677-88. [PMID: 23926430 PMCID: PMC3728269 DOI: 10.2147/ijn.s45429] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Melanoma is the most aggressive type of skin cancer and has very high rates of mortality. An early stage melanoma can be surgically removed, with a survival rate of 99%. However, metastasized melanoma is difficult to cure. The 5-year survival rates for patients with metastasized melanoma are still below 20%. Metastasized melanoma is currently treated by chemotherapy, targeted therapy, immunotherapy and radiotherapy. The outcome of most of the current therapies is far from optimistic. Although melanoma patients with a mutation in the oncogene v-Raf murine sarcoma viral oncogene homolog B1 (BRAF) have an initially higher positive response rate to targeted therapy, the majority develop acquired drug resistance after 6 months of the therapy. To increase treatment efficacy, early diagnosis, more potent pharmacological agents, and more effective delivery systems are urgently needed. Nanotechnology has been extensively studied for melanoma treatment and diagnosis, to decrease drug resistance, increase therapeutic efficacy, and reduce side effects. In this review, we summarize the recent progress on the development of various nanoparticles for melanoma treatment and diagnosis. Several common nanoparticles, including liposome, polymersomes, dendrimers, carbon-based nanoparticles, and human albumin, have been used to deliver chemotherapeutic agents, and small interfering ribonucleic acids (siRNAs) against signaling molecules have also been tested for the treatment of melanoma. Indeed, several nanoparticle-delivered drugs have been approved by the US Food and Drug Administration and are currently in clinical trials. The application of nanoparticles could produce side effects, which will need to be reduced so that nanoparticle-delivered drugs can be safely applied in the clinical setting.
Collapse
Affiliation(s)
- Jiezhong Chen
- School of Biomedical Sciences, University of Queensland, Brisbane, QLD, Australia.
| | | | | | | |
Collapse
|
143
|
Ruggeri BA, Camp F, Miknyoczki S. Animal models of disease: pre-clinical animal models of cancer and their applications and utility in drug discovery. Biochem Pharmacol 2013; 87:150-61. [PMID: 23817077 DOI: 10.1016/j.bcp.2013.06.020] [Citation(s) in RCA: 201] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2013] [Revised: 06/20/2013] [Accepted: 06/21/2013] [Indexed: 01/03/2023]
Abstract
Preclinical models of human cancers are indispensable in the drug discovery and development process for new cancer drugs, small molecules and biologics. They are however imperfect facsimiles of human cancers given the genetic and epigenetic heterogeneity of the latter and the multiplicity of dysregulated survival and growth-regulatory pathways that characterize this spectrum of diseases. This review discusses pre-clinical tumor models - traditional ectopic xenografts, orthotopic xenografts, genetically engineered tumor models, primary human tumorgrafts, and various multi-stage carcinogen-induced tumor models - their advantages, limitations, physiological and pathological relevance. Collectively, these animal models represent a portfolio of test systems that should be utilized at specific stages in the drug discovery process in a pragmatic and hierarchical manner of increasing complexity, physiological relevance, and clinical predictability of the human response. Additionally, evaluating the efficacy of novel therapeutic agents emerging from drug discovery programs in a variety of pre-clinical models can better mimic the heterogeneity of human cancers and also aid in establishing dose levels, dose regimens and drug combinations for use in clinical trials. Nonetheless, despite the sophistication and physiological relevance of these human cancer models (e.g., genetically engineered tumor models and primary human tumografts), the ultimate proof of concept for efficacy and safety of novel oncology therapeutics lies in humans. The judicious interpretation and extrapolation of data derived from these models to humans, and a correspondingly greater emphasis placed on translational medical research in early stage clinical trials, are essential to improve on the current clinical attrition rates for novel oncology therapeutic agents.
Collapse
Affiliation(s)
- Bruce A Ruggeri
- Oncology Discovery & Product Development, Global Branded R & D, Teva Pharmaceuticals, Inc, 145 Brandywine Parkway, West Chester, PA 19380, USA.
| | - Faye Camp
- Oncology Discovery & Product Development, Global Branded R & D, Teva Pharmaceuticals, Inc, 145 Brandywine Parkway, West Chester, PA 19380, USA
| | - Sheila Miknyoczki
- Oncology Discovery & Product Development, Global Branded R & D, Teva Pharmaceuticals, Inc, 145 Brandywine Parkway, West Chester, PA 19380, USA
| |
Collapse
|
144
|
Abstract
PURPOSE OF REVIEW The landscape of medical oncology is filled with approvals of new anticancer agents, the majority of which are targeted agents. This shift in therapies raises multiple challenges including the appearance of new toxicities, the need for biomarkers, the emergence of genomics and the evolution of cancer molecular imaging. RECENT FINDINGS Biopsy of metastatic lesions is slowly becoming a standard of care before the initiation of any therapy. These invasive procedures have been found to be generally well tolerated and are being put to use with the emergence of genomics. Gene sequencing and new imaging techniques are serving the understanding of tumor biology and the search for 'biomarkers' predicting response and resistance to treatment. New clinical trial designs incorporating the 'presumed' biomarkers are guiding patients to specific treatments and have shown outcome improvements. SUMMARY Many questions remain however unanswered and new challenges appear. Intratumor heterogeneity emerges as a brake to personalized cancer care. Relevant targets remain undruggable and costs may hinder progress. Furthermore, technical issues continue to arise. The ultimate goal remains to achieve cure by blocking the multiple pathways of cancer development and proliferation, while sparing the patients the burden of therapy.
Collapse
|
145
|
Peters S, Michielin O, Zimmermann S. Dramatic response induced by vemurafenib in a BRAF V600E-mutated lung adenocarcinoma. J Clin Oncol 2013; 31:e341-4. [PMID: 23733758 DOI: 10.1200/jco.2012.47.6143] [Citation(s) in RCA: 89] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Affiliation(s)
- Solange Peters
- Centre Hospitalier Universitaire Vaudois, Oncology Department, CHUV BH06, Rue du Bugnon 46, 1011 Lausanne, Switzerland
| | | | | |
Collapse
|
146
|
Abstract
Advanced differentiated thyroid cancer (DTC), defined by clinical characteristics including gross extrathyroidal invasion, distant metastases, radioiodine (RAI) resistance, and avidity for 18-fluorodeoxyglucose (positron emission tomography-positive), is found in approximately 10-20% of patients with DTC. Standard therapy (surgery, RAI, TSH suppression with levothyroxine) is ineffective for many of these patients, as is standard chemotherapy. Our understanding of the molecular mechanisms leading to DTC and the transformation to advanced DTC has rapidly evolved over the past 15-20 years. Newer targeted therapy, specifically inhibitors of intracellular kinase signaling pathways, and cooperative multicenter clinical trials have dramatically changed the therapeutic landscape for patients with advanced DTC. In this review focusing on morbidities, molecules, and medicinals, we present a patient with advanced DTC, explore the genetics and molecular biology of advanced DTC, and review evolving therapies for these patients including multikinase inhibitors, selective kinase inhibitors, and combination therapies.
Collapse
Affiliation(s)
- Bryan R Haugen
- University of Colorado School of Medicine, University of Colorado Cancer Center, Aurora, Colorado 80045, USA.
| | | |
Collapse
|
147
|
Scott AJ, Leong S, Messersmith WA, Lieu CH. A moving target: challenges in treating BRAF-mutant colorectal cancer. COLORECTAL CANCER 2013. [DOI: 10.2217/crc.13.23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
SUMMARY Biomarker-oriented clinical studies have shown mounting evidence for improved efficacy with targeted therapy. Unfortunately, single-agent BRAF inhibition has shown limited efficacy in patients with metastatic BRAF-mutant colorectal cancer. Multiple mechanisms for potential BRAF inhibitor resistance demonstrate the complex biology behind tumorigenesis in colorectal cancer and highlight the obstacles of acquired and intrinsic drug resistance in treating this disease. Biomarker-related strategies with potential predictive value for specific molecular anomalies offer the possibility for improved targeted agents and rational combinations. Synthetic lethality gene array has become a major focus in current research for developing such agents. Combination therapy based on molecular profiles may further enhance individualized patient care.
Collapse
Affiliation(s)
- Aaron J Scott
- University of Colorado, MS8117, 12801 E 17th Avenue, Room 8122, Aurora, CO 80024, USA
| | - Stephen Leong
- University of Colorado, MS8117, 12801 E 17th Avenue, Room 8122, Aurora, CO 80024, USA
| | - Wells A Messersmith
- University of Colorado, MS8117, 12801 E 17th Avenue, Room 8122, Aurora, CO 80024, USA
| | - Christopher H Lieu
- University of Colorado, MS8117, 12801 E 17th Avenue, Room 8122, Aurora, CO 80024, USA.
| |
Collapse
|
148
|
Fogliatto G, Gianellini L, Brasca MG, Casale E, Ballinari D, Ciomei M, Degrassi A, De Ponti A, Germani M, Guanci M, Paolucci M, Polucci P, Russo M, Sola F, Valsasina B, Visco C, Zuccotto F, Donati D, Felder E, Pesenti E, Galvani A, Mantegani S, Isacchi A. NMS-E973, a Novel Synthetic Inhibitor of Hsp90 with Activity against Multiple Models of Drug Resistance to Targeted Agents, Including Intracranial Metastases. Clin Cancer Res 2013; 19:3520-32. [DOI: 10.1158/1078-0432.ccr-12-3512] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
149
|
Turajlic S, Ali Z, Yousaf N, Larkin J. Phase I/II RAF kinase inhibitors in cancer therapy. Expert Opin Investig Drugs 2013; 22:739-49. [PMID: 23642225 DOI: 10.1517/13543784.2013.797964] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
INTRODUCTION Aberrant activation of RAF signalling is a frequent finding in human cancers. BRAF is the only RAF family member that is commonly mutated, whilst CRAF and ARAF play important roles in the signal transduction from mutant RAS. BRAF-specific inhibitors have been more effective in the treatment of BRAF-mutant melanoma than BRAF-mutant thyroid and colorectal cancers. AREAS COVERED The review summarises the experience with RAF kinase inhibitors, including efficacy, modes of acquired resistance, and the mechanism behind the progression of pre-malignant RAS-mutant lesions observed with RAF kinase inhibitors. The authors review all the completed and ongoing Phase I or II clinical trials of RAF kinase inhibitors and discuss in detail the rationale behind the combinatorial approaches. EXPERT OPINION The success of RAF kinase inhibitors has demonstrated the necessity of genotype-driven treatment selection for cancer patients. The spectrum of responses in different tumour types is explained by feedback events that are determined by cell lineage. Dissection of these events and the mechanisms of acquired resistance will determine the appropriate combination therapies. Ongoing characterisation of RAS-MAPK regulation in malignant cells may aid the development of novel agents that have greater potency for the inhibition of activated RAF kinase, and lesser propensity for promotion of RAS-mutant tumours.
Collapse
Affiliation(s)
- Samra Turajlic
- Royal Marsden Hospital, Melanoma Unit, London SW3 6JJ, UK
| | | | | | | |
Collapse
|
150
|
Abstract
a substantial proportion of non-small-cell lung cancer (NSCLC), and adenocarcinoma in particular, depends on a so-called 'driver mutation' for their malignant phenotype. This genetic alteration induces and sustains tumorigenesis, and targeting of its protein product can result in growth inhibition, tumor response and increased patient survival. NSCLC can thus be subdivided into clinically relevant molecular subsets. Mutations in EGFR best illustrate the therapeutic relevance of molecular classification. This article reviews the scope of presently known driving molecular alterations, including ROS1, BRAF, KRAS, HER2 and PIK3CA, with a special emphasis on aLK rearrangements, and outlines their potential therapeutic applications.
Collapse
Affiliation(s)
- S Zimmermann
- Department of Oncology, Lausanne University Hospital, Lausanne, Switzerland
| | | |
Collapse
|