1
|
Zheng E, Włodarczyk M, Węgiel A, Osielczak A, Możdżan M, Biskup L, Grochowska A, Wołyniak M, Gajewski D, Porc M, Maryńczak K, Dziki Ł. Navigating through novelties concerning mCRC treatment-the role of immunotherapy, chemotherapy, and targeted therapy in mCRC. Front Surg 2024; 11:1398289. [PMID: 38948479 PMCID: PMC11211389 DOI: 10.3389/fsurg.2024.1398289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Accepted: 05/29/2024] [Indexed: 07/02/2024] Open
Abstract
Over the course of nearly six decades since the inception of initial trials involving 5-FU in the treatment of mCRC (metastatic colorectal cancer), our progressive comprehension of the pathophysiology, genetics, and surgical techniques related to mCRC has paved the way for the introduction of novel therapeutic modalities. These advancements not only have augmented the overall survival but have also positively impacted the quality of life (QoL) for affected individuals. Despite the remarkable progress made in the last two decades in the development of chemotherapy, immunotherapy, and target therapies, mCRC remains an incurable disease, with a 5-year survival rate of 14%. In this comprehensive review, our primary goal is to present an overview of mCRC treatment methods following the latest guidelines provided by the National Comprehensive Cancer Network (NCCN), the American Society of Clinical Oncology (ASCO), and the American Society of Colon and Rectal Surgeons (ASCRS). Emphasis has been placed on outlining treatment approaches encompassing chemotherapy, immunotherapy, targeted therapy, and surgery's role in managing mCRC. Furthermore, our review delves into prospective avenues for developing new therapies, offering a glimpse into the future of alternative pathways that hold potential for advancing the field.
Collapse
Affiliation(s)
- Edward Zheng
- Department of General and Oncological Surgery, Faculty of Medicine, Medical University of Lodz, Lodz, Poland
| | - Marcin Włodarczyk
- Department of General and Oncological Surgery, Faculty of Medicine, Medical University of Lodz, Lodz, Poland
| | - Andrzej Węgiel
- Department of General and Oncological Surgery, Faculty of Medicine, Medical University of Lodz, Lodz, Poland
| | - Aleksandra Osielczak
- Department of General and Oncological Surgery, Faculty of Medicine, Medical University of Lodz, Lodz, Poland
| | - Maria Możdżan
- Department of General and Oncological Surgery, Faculty of Medicine, Medical University of Lodz, Lodz, Poland
| | - Laura Biskup
- Department of General and Oncological Surgery, Faculty of Medicine, Medical University of Lodz, Lodz, Poland
| | - Agata Grochowska
- Department of General and Oncological Surgery, Faculty of Medicine, Medical University of Lodz, Lodz, Poland
| | - Maria Wołyniak
- Department of General and Oncological Surgery, Faculty of Medicine, Medical University of Lodz, Lodz, Poland
- Department of Biostatistics and Translational Medicine, Medical University of Lodz, Lodz, Poland
| | - Dominik Gajewski
- Department of General and Oncological Surgery, Faculty of Medicine, Medical University of Lodz, Lodz, Poland
| | - Mateusz Porc
- Department of General and Oncological Surgery, Faculty of Medicine, Medical University of Lodz, Lodz, Poland
| | - Kasper Maryńczak
- Department of General and Oncological Surgery, Faculty of Medicine, Medical University of Lodz, Lodz, Poland
| | - Łukasz Dziki
- Department of General and Oncological Surgery, Faculty of Medicine, Medical University of Lodz, Lodz, Poland
| |
Collapse
|
2
|
Al-Hujaily EM, Al-Sowayan BS, Alyousef Z, Uddin S, Alammari F. Recruiting Immunity for the Fight against Colorectal Cancer: Current Status and Challenges. Int J Mol Sci 2022; 23:ijms232213696. [PMID: 36430176 PMCID: PMC9697544 DOI: 10.3390/ijms232213696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 10/31/2022] [Accepted: 11/03/2022] [Indexed: 11/09/2022] Open
Abstract
Cancer immunotherapies have changed the landscape of cancer management and improved the standard treatment protocols used in multiple tumors. This has led to significant improvements in progression-free survival and overall survival rates. In this review article, we provide an insight into the major immunotherapeutic methods that are currently under investigation for colorectal cancer (CRC) and their clinical implementations. We emphasize therapies that are based on monoclonal antibodies (mAbs) and adoptive cell therapy, their mechanisms of action, their advantages, and their potential in combination therapy. We also highlight the clinical trials that have demonstrated both the therapeutic efficacy and the toxicities associated with each method. In addition, we summarize emerging targets that are now being evaluated as potential interventions for CRC. Finally, we discuss current challenges and future direction for the cancer immunotherapy field.
Collapse
Affiliation(s)
- Ensaf M. Al-Hujaily
- Department of Blood and Cancer Research, King Abdullah International Medical Research Center, Riyadh 11481, Saudi Arabia
- King Saud Bin Abdulaziz University for Health Sciences, Riyadh 14611, Saudi Arabia
| | - Batla S. Al-Sowayan
- Department of Blood and Cancer Research, King Abdullah International Medical Research Center, Riyadh 11481, Saudi Arabia
- King Saud Bin Abdulaziz University for Health Sciences, Riyadh 14611, Saudi Arabia
| | - Zeyad Alyousef
- Department of Surgery, College of Medicine, King Saud bin Abdulaziz University for Health Sciences, Ministry of National Guard Health Affairs, Riyadh 14611, Saudi Arabia
| | - Shahab Uddin
- Translational Research Institute and Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar
| | - Farah Alammari
- Department of Blood and Cancer Research, King Abdullah International Medical Research Center, Riyadh 11481, Saudi Arabia
- King Saud Bin Abdulaziz University for Health Sciences, Riyadh 14611, Saudi Arabia
- Clinical Laboratory Sciences Department, College of Applied Medical Sciences, King Saud bin Abdulaziz University for Health Sciences, Riyadh 14611, Saudi Arabia
- Correspondence:
| |
Collapse
|
3
|
Abdalbari FH, Telleria CM. The gold complex auranofin: new perspectives for cancer therapy. Discov Oncol 2021; 12:42. [PMID: 35201489 PMCID: PMC8777575 DOI: 10.1007/s12672-021-00439-0] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 10/12/2021] [Indexed: 12/14/2022] Open
Abstract
Advanced stages of cancer are highly associated with short overall survival in patients due to the lack of long-term treatment options following the standard form of care. New options for cancer therapy are needed to improve the survival of cancer patients without disease recurrence. Auranofin is a clinically approved agent against rheumatoid arthritis that is currently enrolled in clinical trials for potential repurposing against cancer. Auranofin mainly targets the anti-oxidative system catalyzed by thioredoxin reductase (TrxR), which protects the cell from oxidative stress and death in the cytoplasm and the mitochondria. TrxR is over-expressed in many cancers as an adaptive mechanism for cancer cell proliferation, rendering it an attractive target for cancer therapy, and auranofin as a potential therapeutic agent for cancer. Inhibiting TrxR dysregulates the intracellular redox state causing increased intracellular reactive oxygen species levels, and stimulates cellular demise. An alternate mechanism of action of auranofin is to mimic proteasomal inhibition by blocking the ubiquitin-proteasome system (UPS), which is critically important in cancer cells to prevent cell death when compared to non-cancer cells, because of its role on cell cycle regulation, protein degradation, gene expression, and DNA repair. This article provides new perspectives on the potential mechanisms used by auranofin alone, in combination with diverse other compounds, or in combination with platinating agents and/or immune checkpoint inhibitors to combat cancer cells, while assessing the feasibility for its repurposing in the clinical setting.
Collapse
Affiliation(s)
- Farah H Abdalbari
- Experimental Pathology Unit, Department of Pathology, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada
| | - Carlos M Telleria
- Experimental Pathology Unit, Department of Pathology, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada.
- Cancer Research Program, Research Institute, McGill University Health Centre, Montreal, QC, Canada.
| |
Collapse
|
4
|
Secombe KR, Van Sebille YZA, Mayo BJ, Coller JK, Gibson RJ, Bowen JM. Diarrhea Induced by Small Molecule Tyrosine Kinase Inhibitors Compared With Chemotherapy: Potential Role of the Microbiome. Integr Cancer Ther 2021; 19:1534735420928493. [PMID: 32493068 PMCID: PMC7273583 DOI: 10.1177/1534735420928493] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Small molecule receptor tyrosine kinase inhibitors (SM-TKIs) are among a group of
targeted cancer therapies, intended to be more specific to cancer cells compared
with treatments, such as chemotherapy, hence reducing adverse events.
Unfortunately, many patients report high levels of diarrhea, the pathogenesis of
which remains under investigation. In this article, we compare the current state
of knowledge of the pathogenesis of chemotherapy-induced diarrhea (CID) in
comparison to SM-TKI–induced diarrhea, and investigate how a similar research
approach in both areas may be beneficial. To this end, we review evidence that
both treatment modalities may interact with the gut microbiome, and as such the
microbiome should be investigated for its ability to reduce the risk of
diarrhea.
Collapse
Affiliation(s)
- Kate R Secombe
- Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia
| | - Ysabella Z A Van Sebille
- UniSA Online, Health Sciences, University of South Australia, Adelaide, South Australia, Australia
| | - Bronwen J Mayo
- Division of Health Sciences, University of South Australia, South Australia, Australia
| | - Janet K Coller
- Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia
| | - Rachel J Gibson
- School of Allied Health Science and Practice, University of Adelaide, South Australia, Australia
| | - Joanne M Bowen
- Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia
| |
Collapse
|
5
|
Mihai MM, Ion A, Giurcăneanu C, Nițipir C, Popa AM, Chifiriuc MC, Popa MI, Říčař J, Popa LG, Sârbu I, Lazăr V. The Impact of Long-Term Antibiotic Therapy of Cutaneous Adverse Reactions to EGFR Inhibitors in Colorectal Cancer Patients. J Clin Med 2021; 10:jcm10153219. [PMID: 34362003 PMCID: PMC8347035 DOI: 10.3390/jcm10153219] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Revised: 07/07/2021] [Accepted: 07/19/2021] [Indexed: 12/24/2022] Open
Abstract
Colorectal cancer (CRC) is an important public health issue, in terms of incidence and mortality, with approximately 1.8 million new cases reported worldwide in 2018. Advancements in understanding pathophysiological key steps in CRC tumorigenesis have led to the development of new targeted therapies such as those based on epidermal growth factor receptor inhibitors (EGFR inhibitors). The cutaneous adverse reactions induced by EGFR inhibitors, particularly papulopustular rash, often require long-term antibiotic treatment with tetracycline agents (mostly minocycline and doxycycline). However, this raises several issues of concern: possible occurrence of gut dysbiosis in already vulnerable CRC patients, selection of highly antibiotic resistant and/or virulent clones, development of adverse reactions related to tetracyclines, interference of antibiotics with the response to oncologic therapy, with a negative impact on disease prognosis etc. In the context of scarce information regarding these issues and controversial opinions regarding the role of tetracyclines in patients under EGFR inhibitors, our aim was to perform a thorough literature review and discuss the main challenges raised by long-term use of tetracyclines in advanced CRC patients receiving this targeted therapy.
Collapse
Affiliation(s)
- Mara Mădălina Mihai
- Department of Oncologic Dermatology, ‘Elias’ Emergency University Hospital, ‘Carol Davila’ University of Medicine and Pharmacy, 020021 Bucharest, Romania; (C.G.); (L.G.P.)
- Department of Dermatology, ‘Elias’ Emergency University Hospital, 011461 Bucharest, Romania
- Department of Microbiology, Faculty of Biology, ICUB—Research Institute of the University of Bucharest, 050657 Bucharest, Romania; (M.-C.C.); (V.L.)
- Correspondence: (M.M.M.); (A.I.); Tel.: +40-74-336-4164 (M.M.M.)
| | - Ana Ion
- Department of Dermatology, ‘Elias’ Emergency University Hospital, 011461 Bucharest, Romania
- Correspondence: (M.M.M.); (A.I.); Tel.: +40-74-336-4164 (M.M.M.)
| | - Călin Giurcăneanu
- Department of Oncologic Dermatology, ‘Elias’ Emergency University Hospital, ‘Carol Davila’ University of Medicine and Pharmacy, 020021 Bucharest, Romania; (C.G.); (L.G.P.)
- Department of Dermatology, ‘Elias’ Emergency University Hospital, 011461 Bucharest, Romania
| | - Cornelia Nițipir
- Department of Oncology, ‘Elias’ Emergency University Hospital, ‘Carol Davila’ University of Medicine and Pharmacy, 020021 Bucharest, Romania; (C.N.); (A.-M.P.)
| | - Ana-Maria Popa
- Department of Oncology, ‘Elias’ Emergency University Hospital, ‘Carol Davila’ University of Medicine and Pharmacy, 020021 Bucharest, Romania; (C.N.); (A.-M.P.)
| | - Mariana-Carmen Chifiriuc
- Department of Microbiology, Faculty of Biology, ICUB—Research Institute of the University of Bucharest, 050657 Bucharest, Romania; (M.-C.C.); (V.L.)
| | - Mircea Ioan Popa
- Department of Microbiology, Faculty of Medicine, ‘Carol Davila’ University of Medicine and Pharmacy, 020021 Bucharest, Romania;
| | - Jan Říčař
- Department of Dermatology and Venereology, Charles University, Medical School and Teaching Hospital Pilsen, 30599 Pilsen, Czech Republic;
| | - Liliana Gabriela Popa
- Department of Oncologic Dermatology, ‘Elias’ Emergency University Hospital, ‘Carol Davila’ University of Medicine and Pharmacy, 020021 Bucharest, Romania; (C.G.); (L.G.P.)
- Department of Dermatology, ‘Elias’ Emergency University Hospital, 011461 Bucharest, Romania
| | - Ionela Sârbu
- Department of Genetics, Faculty of Biology, ICUB—Research Institute of the University of Bucharest, 050657 Bucharest, Romania;
| | - Veronica Lazăr
- Department of Microbiology, Faculty of Biology, ICUB—Research Institute of the University of Bucharest, 050657 Bucharest, Romania; (M.-C.C.); (V.L.)
| |
Collapse
|
6
|
Sur D, Havasi A, Gorzo A, Burz C. A Critical Review of Second-Generation Anti-EGFR Monoclonal Antibodies in Metastatic Colorectal Cancer. Curr Drug Targets 2021; 22:1034-1042. [PMID: 32718285 DOI: 10.2174/1389450121666200727121011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 06/04/2020] [Accepted: 06/09/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND Anti-EGFR monoclonal antibodies (mAbs) have become a relevant solution for the treatment of patients with metastatic colorectal cancer. Current anti-EGFR monoclonal antibodies face a series of problems, including resistance and non-durable response, and RAS and BRAF mutations serve as exclusion criteria for treatment with anti-EGFR mAbs. Advances in molecular tumor profiling and information on subsequent pathways responsible for disease progression and drug resistance helped develop a new generation of anti-EGFR mAbs. These second-generation mAbs have been developed to overcome existing resistance mechanisms and to limit common side effects. For the moment, existing literature suggests that these novel anti-EGFR mAbs are far from finding their way to clinical practice soon. OBJECTIVE In this review, we summarize and evaluate current data regarding ongoing research and completed clinical trials for different second-generation anti-EGFR monoclonal antibodies. CONCLUSION Anti-EGFR mAbs exhibit efficacy in advanced colorectal cancer, but second-generation mAbs failed to prove their benefit in the treatment of metastatic colorectal cancer. Understanding the biological basis of primary and acquired drug resistance could allow scientists to design better clinical trials and develop improved second-generation mAbs.
Collapse
Affiliation(s)
- Daniel Sur
- Department of Medical Oncology, Faculty of Medicine, "Iuliu Hatieganu" University of Medicine and Pharmacy, Cluj- Napoca, Romania
| | - Andrei Havasi
- Department of Medical Oncology, "Ion Chiricuta" Oncology Institute, Cluj-Napoca, Romania
| | - Alecsandra Gorzo
- Department of Medical Oncology, "Ion Chiricuta" Oncology Institute, Cluj-Napoca, Romania
| | - Claudia Burz
- Department of Medical Oncology, "Ion Chiricuta" Oncology Institute, Cluj-Napoca, Romania
| |
Collapse
|
7
|
Yang HL, Liu HW, Shrestha S, Thiyagarajan V, Huang HC, Hseu YC. Antrodia salmonea induces apoptosis and enhances cytoprotective autophagy in colon cancer cells. Aging (Albany NY) 2021; 13:15964-15989. [PMID: 34031264 PMCID: PMC8266357 DOI: 10.18632/aging.203019] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 04/05/2021] [Indexed: 04/07/2023]
Abstract
A traditional Chinese medicinal fungus, Antrodia salmonea (AS), with antioxidant properties is familiar in Taiwan but anti-cancer activity of AS in human colon cancer is ambiguous. Hence, we explored the anti-cancer activity of AS in colon cancer cells. 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay revealed that AS showed a remarkable effect on cell viability in colon cancer cells; SW620, HCT116, and HT29. Annexin V/propidium iodide (PI) stained cells indicated that AS induced both early/late apoptosis in SW620 cells. Additionally, cells treated with AS induced caspase-3 activation, poly (ADP-ribose) polymerase (PARP) cleavage, mitochondrial dysfunction, and Bcl-2 associated X (Bax)/B-cell lymphoma (Bcl-2) dysregulation. Microtubule- associated protein 1A/1B-light chain 3B (LC3-II) accumulation, sequestosome 1 (p62/SQSTM1) activation, autophagy related 4B cysteine peptidase (ATG4B) inactivation, acidic vesicular organelles (AVOs) formation, and Beclin-1/Bcl-2 dysregulation revealed that AS-induced autophagy. Interestingly, cells pretreated with 3-methyladenine (3-MA) strengthened AS-induced caspase-3/apoptosis. Suppression of apoptosis by z-Val-Ala-Asp fluoromethyl ketone (Z-VAD-FMK) did not however block AS-induced autophagy, suggesting that autophagy was not attenuated by the AS-induced apoptosis. Application of N-acetylcysteine (NAC) prevented AS-induced cell death, caspase-3 activation, LC3-II accumulation, and AVOs formation, indicating that AS-induced apoptosis and autophagy was mediated by reactive oxygen species (ROS). Furthermore, AS-induced cytoprotective autophagy and apoptosis through extracellular signal-regulated kinase (ERK) signaling cascades. Moreover, in vivo data disclosed that AS inhibited colitis-associated tumorigenesis in azoxymethane (AOM)-dextran sodium sulphate (DSS)-treated mice. For the first time, we report the anti-cancer properties of this potentially advantageous mushroom for the treatment of human colon cancer.
Collapse
Affiliation(s)
- Hsin-Ling Yang
- Institute of Nutrition, College of Health Care, China Medical University, Taichung 40402, Taiwan
| | - Hui-Wen Liu
- Institute of Nutrition, College of Health Care, China Medical University, Taichung 40402, Taiwan
| | - Sirjana Shrestha
- Institute of Nutrition, College of Health Care, China Medical University, Taichung 40402, Taiwan
| | | | - Hui-Chi Huang
- Department of Chinese Pharmaceutical Sciences and Chinese Medicine Resources, College of Chinese Medicine, China Medical University, Taichung 40402, Taiwan
| | - You-Cheng Hseu
- Department of Cosmeceutics, College of Pharmacy, China Medical University, Taichung 40402, Taiwan
- Department of Health and Nutrition Biotechnology, Asia University, Taichung 41354, Taiwan
- Chinese Medicine Research Center, China Medical University, Taichung 40402, Taiwan
| |
Collapse
|
8
|
Kumar CBP, Raghu MS, Prathibha BS, Prashanth MK, Kanthimathi G, Kumar KY, Parashuram L, Alharthi FA. Discovery of a novel series of substituted quinolines acting as anticancer agents and selective EGFR blocker: Molecular docking study. Bioorg Med Chem Lett 2021; 44:128118. [PMID: 34015505 DOI: 10.1016/j.bmcl.2021.128118] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 05/09/2021] [Accepted: 05/13/2021] [Indexed: 11/18/2022]
Abstract
A Ta2O5-anchored-piperidine-4-carboxylic acid (PPCA) nanoparticle has been synthesized and characterized. It was then used as a highly effective nanocatalyst for the synthesis of quinolin-2(1H)-one derivatives through CO bond functionalization. The special advantage of this heterogeneous solid catalyst is the reusability of the catalyst for up to five cycles without any noticeable reduction in product yields. In comparison, healthy reaction profiles, wide substrate scope, excellent yields and easy workup conditions are the notable highlights of this approach. All the compounds were tested for their anticancer activity against MCF-7 (human breast), HepG2 (human liver), HCT116 (human colorectal), and PC-3 (human prostate) cancer cell lines with the MTT assay. All the compounds were shown to have moderate to good inhibitory effects on tested cancer cell lines. Besides, compounds 5b, 5c and 5d showed good selectivity against epidermal growth factor receptor-tyrosine kinase (EGFR-TK). Molecular docking results showed that active compounds showed a good affinity towards EGFR kinase (PDB ID: 6V6O) by forming two hydrogen bonds with Cys-797 and Tyr-801. All the compounds were screened for computational ADMET and Lipinski analysis.
Collapse
Affiliation(s)
- C B Pradeep Kumar
- Department of Chemistry, Malnad College of Engineering, Hassan 573 202, India
| | - M S Raghu
- Department of Chemistry, New Horizon College of Engineering, Bengaluru 560 103, India
| | - B S Prathibha
- Department of Chemistry, B N M Institute of Technology, Bengaluru 560 070, India
| | - M K Prashanth
- Department of Chemistry, B N M Institute of Technology, Bengaluru 560 070, India.
| | - G Kanthimathi
- Department of Chemistry, Ramco Institute of Technology, Rajapalayam, Tamilnadu, 626117, India
| | - K Yogesh Kumar
- Department of Chemistry, School of Engineering and Technology, Jain University, Ramanagara 562 112, India
| | - L Parashuram
- Department of Chemistry, New Horizon College of Engineering, Bengaluru 560 103, India
| | - Fahad A Alharthi
- Department of Chemistry, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| |
Collapse
|
9
|
Navigating metastatic colorectal treatment options in the USA: a survey of patient acceptance of skin toxicities associated with Vectibix. Support Care Cancer 2021; 29:6731-6740. [PMID: 33973081 PMCID: PMC8464558 DOI: 10.1007/s00520-021-06134-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 03/04/2021] [Indexed: 11/16/2022]
Abstract
Abstract Purpose To understand the extent to which metastatic colorectal cancer (mCRC) patients receive education on the prevention and management associated with skin rash following Vectibix treatment. Furthermore, to investigate how this adverse event affects a patient’s quality of life (QoL) and influences their treatment decisions. Methods A cross-sectional survey was administered to 200 mCRC patients (100 Vectibix users and 100 Vectibix non-users). After excluding respondents who had used cetuximab, 61 Vectibix users and 56 Vectibix non-users remained. Results Most Vectibix users (79%) experienced a skin rash in response to treatment of which 65% considered the rash moderate, 27% mild, and 8% severe. Vectibix users generally felt they were adequately informed about the rash (83%), with the most common messages received related to sun protection. However, sunscreen was used by only 42% of patients prior to rash and 60% of patients following the appearance of rash. The use of oral antibiotics was low prior to rash (21%) and following rash (46%). Among patients experiencing a rash within the past week (n=16), 75% reported the rash had a large negative impact on their QoL based on the Dermatology Life Quality Index. Conclusion There was a disconnect between patients feeling they were adequately informed and use of prevention and management strategies such as sun protection. This suggests a gap in patient education and adoption currently exists on management strategies both prior to and following the appearance of rash. Given the negative impact that skin toxicity has on the patient’s quality of life, it is essential that patients receive and subsequently utilize all information that can minimize rash severity.
Collapse
|
10
|
Moradi-Marjaneh R, Asgharzadeh F, Khordad E, Marjaneh MM. The Clinical Impact of Quantitative Cell-free DNA, KRAS, and BRAF Mutations on Response to Anti-EGFR Treatment in Patients with Metastatic Colorectal Cancer. Curr Pharm Des 2021; 27:942-952. [PMID: 33030125 DOI: 10.2174/1381612826666201007163116] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Accepted: 08/31/2020] [Indexed: 11/22/2022]
Abstract
Colorectal cancer (CRC) is one of the most common leading causes of cancer death in the world. Although EGFR inhibitors have established efficacy in metastatic colorectal cancer (mCRC), some patients do not respond to this treatment. The EGFR inhibitors' failure and acquired resistance are partly due to KRAS and BRAF mutations. Thus, prognostic biomarkers that help to select eligible patients are highly in demand. To improve patient selection, assessment of mutational status in circulating cell free DNA (cfDNA), which possibly represents the dynamicity of tumor genetic status better than tumor tissue, could be advantageous. This review summarizes the current knowledge of the prognostic value of cfDNA in patients with mCRC treated with EGFR inhibitors with emphasis on the clinical importance of identification of KRAS and BRAF mutations.
Collapse
Affiliation(s)
- Reyhaneh Moradi-Marjaneh
- Department of Basic Sciences, Faculty of Medicine, Gonabad University of Medical Sciences, Gonabad, Iran
| | - Fereshteh Asgharzadeh
- Department of Physiology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Elnaz Khordad
- Department of Physiology, School of Paramedical Sciences, Torbat Heydariyeh University of Medical Sciences, Torbat Heydariyeh, Iran
| | | |
Collapse
|
11
|
Yorulmaz A, Yalcin B. Panitumumab-Induced Paronychia: A Case Report and a Brief Review of the Literature. Skin Appendage Disord 2021; 7:123-126. [PMID: 33796558 DOI: 10.1159/000512036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 10/04/2020] [Indexed: 11/19/2022] Open
Abstract
Panitumumab is a recombinant, fully humanized IgG2 monoclonal antibody targeting the epidermal growth factor receptor (EGFR). Panitumumab is indicated for patients with metastatic colorectal cancer with progressive refractory disease. Targeted therapies are well known to be well tolerated; however, they may induce toxicities that are distinct from those of classical chemotherapeutic agents. For instance, EGFR inhibitors (EGFRIs) are associated with some specific dermatological adverse effects, one of which is nail toxicity. Since panitumumab is fully humanized, unlike most of the other EGFRIs, it has been reported to have reduced incidence of adverse reactions. Nail-related adverse effects are frequently observed with EGFRIs. A literature search has yielded a list of reviews describing panitumumab-induced nail toxicity. However, as far as we know, there is no case report detailing this adverse effect of panitumumab. Here, we present a case of panitumumab-induced paronychia in a 60-year-old woman with metastatic colon cancer. With this case report, we would like to review the literature and discuss the possible underlying mechanisms of this condition.
Collapse
Affiliation(s)
- Ahu Yorulmaz
- Department of Dermatology, Ankara Bilkent City Hospital, Ankara, Turkey
| | - Basak Yalcin
- Department of Dermatology, Ankara Bilkent City Hospital, Ankara, Turkey
| |
Collapse
|
12
|
Freire Boullosa L, Van Loenhout J, Flieswasser T, De Waele J, Hermans C, Lambrechts H, Cuypers B, Laukens K, Bartholomeus E, Siozopoulou V, De Vos WH, Peeters M, Smits ELJ, Deben C. Auranofin reveals therapeutic anticancer potential by triggering distinct molecular cell death mechanisms and innate immunity in mutant p53 non-small cell lung cancer. Redox Biol 2021; 42:101949. [PMID: 33812801 PMCID: PMC8113045 DOI: 10.1016/j.redox.2021.101949] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 03/12/2021] [Accepted: 03/15/2021] [Indexed: 12/22/2022] Open
Abstract
Auranofin (AF) is an FDA-approved antirheumatic drug with anticancer properties that acts as a thioredoxin reductase 1 (TrxR) inhibitor. The exact mechanisms through which AF targets cancer cells remain elusive. To shed light on the mode of action, this study provides an in-depth analysis on the molecular mechanisms and immunogenicity of AF-mediated cytotoxicity in the non-small cell lung cancer (NSCLC) cell line NCI–H1299 (p53 Null) and its two isogenic derivates with mutant p53 R175H or R273H accumulation. TrxR is highly expressed in a panel of 72 NSCLC patients, making it a valid druggable target in NSCLC for AF. The presence of mutant p53 overexpression was identified as an important sensitizer for AF in (isogenic) NSCLC cells as it was correlated with reduced thioredoxin (Trx) levels in vitro. Transcriptome analysis revealed dysregulation of genes involved in oxidative stress response, DNA damage, granzyme A (GZMA) signaling and ferroptosis. Although functionally AF appeared a potent inhibitor of GPX4 in all NCI–H1299 cell lines, the induction of lipid peroxidation and consequently ferroptosis was limited to the p53 R273H expressing cells. In the p53 R175H cells, AF mainly induced large-scale DNA damage and replication stress, leading to the induction of apoptotic cell death rather than ferroptosis. Importantly, all cell death types were immunogenic since the release of danger signals (ecto-calreticulin, ATP and HMGB1) and dendritic cell maturation occurred irrespective of (mutant) p53 expression. Finally, we show that AF sensitized cancer cells to caspase-independent natural killer cell-mediated killing by downregulation of several key targets of GZMA. Our data provides novel insights on AF as a potent, clinically available, off-patent cancer drug by targeting mutant p53 cancer cells through distinct cell death mechanisms (apoptosis and ferroptosis). In addition, AF improves the innate immune response at both cytostatic (natural killer cell-mediated killing) and cytotoxic concentrations (dendritic cell maturation).
Collapse
Affiliation(s)
- Laurie Freire Boullosa
- Center for Oncological Research (CORE), Integrated Personalized & Precision Oncology Network (IPPON), University of Antwerp, Wilrijk, Belgium.
| | - Jinthe Van Loenhout
- Center for Oncological Research (CORE), Integrated Personalized & Precision Oncology Network (IPPON), University of Antwerp, Wilrijk, Belgium
| | - Tal Flieswasser
- Center for Oncological Research (CORE), Integrated Personalized & Precision Oncology Network (IPPON), University of Antwerp, Wilrijk, Belgium
| | - Jorrit De Waele
- Center for Oncological Research (CORE), Integrated Personalized & Precision Oncology Network (IPPON), University of Antwerp, Wilrijk, Belgium
| | - Christophe Hermans
- Center for Oncological Research (CORE), Integrated Personalized & Precision Oncology Network (IPPON), University of Antwerp, Wilrijk, Belgium; Department of Pathology, Antwerp University Hospital, Edegem, Belgium
| | - Hilde Lambrechts
- Center for Oncological Research (CORE), Integrated Personalized & Precision Oncology Network (IPPON), University of Antwerp, Wilrijk, Belgium
| | - Bart Cuypers
- Adrem Data Lab, Department of Computer Science, University of Antwerp, Antwerp, Belgium; Molecular Parasitology Unit, Department of Biomedical Sciences, Institute of Tropical Medicine, Antwerp, Belgium
| | - Kris Laukens
- Adrem Data Lab, Department of Computer Science, University of Antwerp, Antwerp, Belgium
| | - Esther Bartholomeus
- Department of Medical Genetics, University of Antwerp, Antwerp University Hospital, Edegem, Belgium
| | | | - Winnok H De Vos
- Laboratory of Cell Biology and Histology, Department of Veterinary Sciences, University of Antwerp, Wilrijk, Belgium
| | - Marc Peeters
- Center for Oncological Research (CORE), Integrated Personalized & Precision Oncology Network (IPPON), University of Antwerp, Wilrijk, Belgium; Department of Oncology, Multidisciplinary Oncological Center Antwerp, Antwerp University Hospital, Edegem, Belgium
| | - Evelien L J Smits
- Center for Oncological Research (CORE), Integrated Personalized & Precision Oncology Network (IPPON), University of Antwerp, Wilrijk, Belgium; Center for Cell Therapy and Regenerative Medicine, Antwerp University Hospital, Edegem, Belgium
| | - Christophe Deben
- Center for Oncological Research (CORE), Integrated Personalized & Precision Oncology Network (IPPON), University of Antwerp, Wilrijk, Belgium
| |
Collapse
|
13
|
Lisby AN, Flickinger JC, Bashir B, Weindorfer M, Shelukar S, Crutcher M, Snook AE, Waldman SA. GUCY2C as a biomarker to target precision therapies for patients with colorectal cancer. EXPERT REVIEW OF PRECISION MEDICINE AND DRUG DEVELOPMENT 2021; 6:117-129. [PMID: 34027103 DOI: 10.1080/23808993.2021.1876518] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Introduction Colorectal cancer (CRC) is one of the most-deadly malignancies worldwide. Current therapeutic regimens for CRC patients are relatively generic, based primarily on disease type and stage, with little variation. As the field of molecular oncology advances, so too must therapeutic management of CRC. Understanding molecular heterogeneity has led to a new-found promotion for precision therapy in CRC; underlining the diversity of molecularly targeted therapies based on individual tumor characteristics. Areas covered We review current approaches for the treatment of CRC and discuss the potential of precision therapy in advanced CRC. We highlight the utility of the intestinal protein guanylyl cyclase C (GUCY2C), as a multi-purpose biomarker and unique therapeutic target in CRC. Here, we summarize current GUCY2C-targeted approaches for treatment of CRC. Expert opinion The GUCY2C biomarker has multi-faceted utility in medicine. Developmental investment of GUCY2C as a diagnostic and therapeutic biomarker offers a variety of options taking the molecular characteristics of cancer into account. From GUCY2C-targeted therapies, namely cancer vaccines, CAR-T cells, and monoclonal antibodies, to GUCY2C agonists for chemoprevention in those who are at high risk for developing colorectal cancer, the utility of this protein provides many avenues for exploration with significance in the field of precision medicine.
Collapse
Affiliation(s)
- Amanda N Lisby
- Department of Pharmacology and Experimental Therapeutics, Thomas Jefferson University, Philadelphia, PA 19107, United States
| | - John C Flickinger
- Department of Pharmacology and Experimental Therapeutics, Thomas Jefferson University, Philadelphia, PA 19107, United States
| | - Babar Bashir
- Department of Medical Oncology, Thomas Jefferson University, Philadelphia, PA 19107, United States
| | - Megan Weindorfer
- Department of Pharmacology and Experimental Therapeutics, Thomas Jefferson University, Philadelphia, PA 19107, United States
| | - Sanjna Shelukar
- Department of Pharmacology and Experimental Therapeutics, Thomas Jefferson University, Philadelphia, PA 19107, United States
| | - Madison Crutcher
- Department of Surgery, Thomas Jefferson University, Philadelphia, PA 19107, United States
| | - Adam E Snook
- Department of Pharmacology and Experimental Therapeutics, Thomas Jefferson University, Philadelphia, PA 19107, United States
| | - Scott A Waldman
- Department of Pharmacology and Experimental Therapeutics, Thomas Jefferson University, Philadelphia, PA 19107, United States
| |
Collapse
|
14
|
Pei J, Juniper G, van den Berg NS, Nisho N, Broadt T, Welch AR, Yi GS, Raymundo RC, Chirita SU, Lu G, Krishnan G, Lee YJ, Kapoor S, Zhou Q, Colevas AD, Lui NS, Poultsides GA, Li G, Zinn KR, Rosenthal EL. Safety and Stability of Antibody-Dye Conjugate in Optical Molecular Imaging. Mol Imaging Biol 2021; 23:109-116. [PMID: 32880818 PMCID: PMC9398032 DOI: 10.1007/s11307-020-01536-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 08/03/2020] [Accepted: 08/20/2020] [Indexed: 01/06/2023]
Abstract
PURPOSE The development of molecularly targeted tracers is likely to improve the accuracy of diagnostic, screening, and therapeutic tools. Despite the many therapeutic antibodies that are FDA-approved with known toxicity, only a limited number of antibody-dye conjugates have been introduced to the clinic. Thorough evaluation of the safety, stability, and pharmacokinetics of antibody conjugates in the clinical setting compared with their parental components could accelerate the clinical approval of antibodies as agents for molecular imaging. Here we investigate the safety and stability of a near-infrared fluorescent dye (IRDye800CW) conjugated panitumumab, an approved therapeutic antibody, and report on the product stability, pharmacokinetics, adverse events, and QTc interval changes in patients. PROCEDURES Panitumumab-IRDye800CW was made under good manufacturing practice (GMP) conditions in a single batch on March 26, 2014, and then evaluated over 4.5 years at 0, 3, and 6 months, and then at 6-month intervals thereafter. We conducted early phase trials in head and neck, lung, pancreas, and brain cancers with panitumumab-IRDye800CW. Eighty-one patients scheduled to undergo standard-of-care surgery were infused with doses between 0.06 to 2.83 mg/kg of antibody. Patient ECGs, blood samples, and adverse events were collected over 30-day post-infusion for analysis. RESULTS Eighty-one patients underwent infusion of the study drug at a range of doses. Six patients (7.4 %) experienced an adverse event that was considered potentially related to the drug. The most common event was a prolonged QTc interval which occurred in three patients (3.7 %). Panitumumab-IRDye800CW had two OOS results at 42 and 54 months while meeting all other stability testing criteria. CONCLUSIONS Panitumumab-IRDye800CW was safe and stable to administer over a 54-month window with a low rate of adverse events (7.4 %) which is consistent with the rate associated with panitumumab alone. This data supports re-purposing therapeutic antibodies as diagnostic imaging agents with limited preclinical toxicology studies.
Collapse
Affiliation(s)
- Jacqueline Pei
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, 900 Blake Wilbur Drive, Stanford, CA, 94305, USA
| | - Georgina Juniper
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, 900 Blake Wilbur Drive, Stanford, CA, 94305, USA
| | - Nynke S van den Berg
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, 900 Blake Wilbur Drive, Stanford, CA, 94305, USA
| | - Naoki Nisho
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, 900 Blake Wilbur Drive, Stanford, CA, 94305, USA
| | - Trevor Broadt
- Biopharmaceutical Development Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Anthony R Welch
- Biological Resources Branch/DTP/DCTD, National Cancer Institute, Frederick, MD, USA
| | - Grace S Yi
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, 900 Blake Wilbur Drive, Stanford, CA, 94305, USA
| | - Roan C Raymundo
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, 900 Blake Wilbur Drive, Stanford, CA, 94305, USA
| | - Stefania U Chirita
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, 900 Blake Wilbur Drive, Stanford, CA, 94305, USA
| | - Guolan Lu
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, 900 Blake Wilbur Drive, Stanford, CA, 94305, USA
| | - Giri Krishnan
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, 900 Blake Wilbur Drive, Stanford, CA, 94305, USA
| | - Yu-Jin Lee
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, 900 Blake Wilbur Drive, Stanford, CA, 94305, USA
| | - Shrey Kapoor
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, 900 Blake Wilbur Drive, Stanford, CA, 94305, USA
| | - Quan Zhou
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, 900 Blake Wilbur Drive, Stanford, CA, 94305, USA
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, USA
| | - A Dimitrios Colevas
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, 900 Blake Wilbur Drive, Stanford, CA, 94305, USA
| | - Natalie S Lui
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - George A Poultsides
- Department of Surgery, Section of Surgical Oncology, Stanford University School of Medicine, Stanford, CA, USA
| | - Gordon Li
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Kurt R Zinn
- Department of Radiology, Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, USA
| | - Eben L Rosenthal
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, 900 Blake Wilbur Drive, Stanford, CA, 94305, USA.
| |
Collapse
|
15
|
Coleman EL, Olamiju B, Leventhal JS. Potentially life‑threatening severe cutaneous adverse reactions associated with tyrosine kinase inhibitors (Review). Oncol Rep 2020; 45:891-898. [PMID: 33650659 DOI: 10.3892/or.2020.7911] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 10/21/2020] [Indexed: 11/05/2022] Open
Abstract
Tyrosine kinase inhibitors (TKIs) have emerged as a new frontier of cancer therapy. These agents include inhibitors of epidermal growth factor receptor (EGFR), human epidermal growth factor receptor 2 (HER2), BRAF, mitogen‑activated protein kinase kinase (also referred to as MEK), bcr‑abl, c‑KIT, platelet‑derived growth factor (PDGFR), fibroblast growth factor receptor (FGFR), anaplastic lymphoma kinase (ALK) and vascular endothelial growth factor (VEGF). Along with the evolving applications of TKIs, there has been an increased recognition of the breadth of potential cutaneous toxicities to these agents. In this review, we provide an overview of potentially life‑threatening severe cutaneous adverse reactions (SCARs) that may occur during therapy with TKIs. These toxicities include Stevens‑Johnson Syndrome (SJS), toxic epidermal necrolysis (TEN), drug reaction with eosinophilia and systemic symptoms (DRESS), and acute generalized exanthematous pustulosis (AGEP).
Collapse
|
16
|
Leopold AV, Verkhusha VV. Light control of RTK activity: from technology development to translational research. Chem Sci 2020; 11:10019-10034. [PMID: 33209247 PMCID: PMC7654314 DOI: 10.1039/d0sc03570j] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Accepted: 08/30/2020] [Indexed: 12/11/2022] Open
Abstract
Inhibition of receptor tyrosine kinases (RTKs) by small molecule inhibitors and monoclonal antibodies is used to treat cancer. Conversely, activation of RTKs with their ligands, including growth factors and insulin, is used to treat diabetes and neurodegeneration. However, conventional therapies that rely on injection of RTK inhibitors or activators do not provide spatiotemporal control over RTK signaling, which results in diminished efficiency and side effects. Recently, a number of optogenetic and optochemical approaches have been developed that allow RTK inhibition or activation in cells and in vivo with light. Light irradiation can control RTK signaling non-invasively, in a dosed manner, with high spatio-temporal precision, and without the side effects of conventional treatments. Here we provide an update on the current state of the art of optogenetic and optochemical RTK technologies and the prospects of their use in translational studies and therapy.
Collapse
Affiliation(s)
- Anna V Leopold
- Medicum , Faculty of Medicine , University of Helsinki , Helsinki 00290 , Finland
| | - Vladislav V Verkhusha
- Medicum , Faculty of Medicine , University of Helsinki , Helsinki 00290 , Finland
- Department of Anatomy and Structural Biology and Gruss-Lipper Biophotonics Center , Albert Einstein College of Medicine , Bronx , NY 10461 , USA .
| |
Collapse
|
17
|
Kattan WE, Hancock JF. RAS Function in cancer cells: translating membrane biology and biochemistry into new therapeutics. Biochem J 2020; 477:2893-2919. [PMID: 32797215 PMCID: PMC7891675 DOI: 10.1042/bcj20190839] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 07/20/2020] [Accepted: 07/22/2020] [Indexed: 02/07/2023]
Abstract
The three human RAS proteins are mutated and constitutively activated in ∼20% of cancers leading to cell growth and proliferation. For the past three decades, many attempts have been made to inhibit these proteins with little success. Recently; however, multiple methods have emerged to inhibit KRAS, the most prevalently mutated isoform. These methods and the underlying biology will be discussed in this review with a special focus on KRAS-plasma membrane interactions.
Collapse
Affiliation(s)
- Walaa E. Kattan
- Department of Integrative Biology and Pharmacology, McGovern Medical School University of Texas Health Science Center at Houston, TX 77030, USA
- The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, TX 77030, USA
| | - John F. Hancock
- Department of Integrative Biology and Pharmacology, McGovern Medical School University of Texas Health Science Center at Houston, TX 77030, USA
- The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, TX 77030, USA
| |
Collapse
|
18
|
Chuang SC, Huang CW, Chen YT, Ma CJ, Tsai HL, Chang TK, Su WC, Hsu WH, Kuo CH, Wang JY. Effect of KRAS and NRAS mutations on the prognosis of patients with synchronous metastatic colorectal cancer presenting with liver-only and lung-only metastases. Oncol Lett 2020; 20:2119-2130. [PMID: 32782529 PMCID: PMC7400335 DOI: 10.3892/ol.2020.11795] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Accepted: 03/26/2020] [Indexed: 12/24/2022] Open
Abstract
It has been reported that 20–25% of patients with colorectal cancer (CRC) have metastases at the time of diagnosis. Liver and lung are the most common metastatic sites. The aim of the present study was to investigate the association of KRAS and NRAS mutations with clinicopathological features and prognosis of patients with initial liver-metastasis only (LiM-only) or lung-metastasis only (LuM-only) metastatic CRC (mCRC). Overall, 166 patients with CRC with initial LiM-only (n=124) and LuM-only (n=42) were retrospectively analyzed from January 2014 to December 2017. The median follow-up time was 19.2 months (1.0–57.1 months). Patient characteristics at diagnosis were collected. Genomic DNA was isolated from frozen primary CRC tissues for targeting KRAS and NRAS. Patients with LuM-only were significantly older compared with those with LiM-only (65.5 vs. 61.5 years; P=0.05). There was no significant differences between the LiM-only and LuM-only groups in terms of sex, location of the primary tumor, serum carcinoembryonic antigen level, histological grade and RAS mutation status. KRAS mutations were detected in 43 (41.0%) patients with LiM-only and 13 (35.1%) patients with LuM-only. The overall survival time (OS) of LuM-only was more favorable compared with that of patients with LiM-only (44.5 vs. 24.7 months); however, there was no significant difference (P=0.095). The progression-free survival (PFS) and OS in the RAS wild-type group were significantly improved compared with the RAS mutant cohorts (P=0.004 and P=0.031, respectively) in the LiM-only group. In patients with stage IV CRC, those with synchronous LiM-only mCRC had a higher incidence of metastasis but a less favorable PFS and OS compared with patients with LuM-only. RAS mutation status exhibited a significant association with the survival outcome in patients with LiM-only mCRC.
Collapse
Affiliation(s)
- Shih-Chang Chuang
- Division of General and Digestive Surgery, Department of Surgery; Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan, R.O.C.,Department of Surgery, Faculty of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan, R.O.C
| | - Ching-Wen Huang
- Department of Surgery, Faculty of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan, R.O.C.,Division of Colorectal Surgery, Department of Surgery, Kaohsiung Medical University, Kaohsiung 807, Taiwan, R.O.C
| | - Yi-Ting Chen
- Department of Pathology; Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan, R.O.C.,Department of Pathology, Faculty of Medicine, College of Medicine, Kaohsiung 807, Taiwan, R.O.C.,Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan, R.O.C
| | - Cheng-Jen Ma
- Division of General and Digestive Surgery, Department of Surgery; Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan, R.O.C
| | - Hsiang-Lin Tsai
- Department of Surgery, Faculty of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan, R.O.C.,Division of Colorectal Surgery, Department of Surgery, Kaohsiung Medical University, Kaohsiung 807, Taiwan, R.O.C
| | - Tsung-Kun Chang
- Division of Colorectal Surgery, Department of Surgery, Kaohsiung Medical University, Kaohsiung 807, Taiwan, R.O.C
| | - Wei-Chih Su
- Division of Colorectal Surgery, Department of Surgery, Kaohsiung Medical University, Kaohsiung 807, Taiwan, R.O.C
| | - Wen-Hung Hsu
- Division of Gastroenterology, Department of Internal Medicine, Kaohsiung Medical University Hospital and Kaohsiung Medical University, Kaohsiung 807, Taiwan, R.O.C
| | - Chao-Hung Kuo
- Division of Gastroenterology, Department of Internal Medicine, Kaohsiung Medical University Hospital and Kaohsiung Medical University, Kaohsiung 807, Taiwan, R.O.C
| | - Jaw-Yuan Wang
- Department of Surgery, Faculty of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan, R.O.C.,Division of Colorectal Surgery, Department of Surgery, Kaohsiung Medical University, Kaohsiung 807, Taiwan, R.O.C.,Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan, R.O.C.,Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung 807, Taiwan, R.O.C.,Cohort Research Center, College of Medicine, Kaohsiung 807, Taiwan, R.O.C.,Center for Cancer Research, Kaohsiung Medical University, Kaohsiung 807, Taiwan, R.O.C
| |
Collapse
|
19
|
Tan Z, Zhu J, Stemmer PM, Sun L, Yang Z, Schultz K, Gaffrey MJ, Cesnik AJ, Yi X, Hao X, Shortreed MR, Shi T, Lubman DM. Comprehensive Detection of Single Amino Acid Variants and Evaluation of Their Deleterious Potential in a PANC-1 Cell Line. J Proteome Res 2020; 19:1635-1646. [PMID: 32058723 DOI: 10.1021/acs.jproteome.9b00840] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Identifying single amino acid variants (SAAVs) in cancer is critical for precision oncology. Several advanced algorithms are now available to identify SAAVs, but attempts to combine different algorithms and optimize them on large data sets to achieve a more comprehensive coverage of SAAVs have not been implemented. Herein, we report an expanded detection of SAAVs in the PANC-1 cell line using three different strategies, which results in the identification of 540 SAAVs in the mass spectrometry data. Among the set of 540 SAAVs, 79 are evaluated as deleterious SAAVs based on analysis using the novel AssVar software in which one of the driver mutations found in each protein of KRAS, TP53, and SLC37A4 is further validated using independent selected reaction monitoring (SRM) analysis. Our study represents the most comprehensive discovery of SAAVs to date and the first large-scale detection of deleterious SAAVs in the PANC-1 cell line. This work may serve as the basis for future research in pancreatic cancer and personal immunotherapy and treatment.
Collapse
Affiliation(s)
- Zhijing Tan
- Department of Surgery, The University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Jianhui Zhu
- Department of Surgery, The University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Paul M Stemmer
- Institute of Environmental Health Sciences, Wayne State University, Detroit, Michigan 48202, United States
| | - Liangliang Sun
- Department of Chemistry, Michigan State University, 578 South Shaw Lane, East Lansing, Michigan 48824, United States
| | - Zhichang Yang
- Department of Chemistry, Michigan State University, 578 South Shaw Lane, East Lansing, Michigan 48824, United States
| | - Kendall Schultz
- Integrative Omics Group, Biological Sciences Division, Pacific Northwest National Laboratory, Richland, Washington 99352, United States
| | - Matthew J Gaffrey
- Integrative Omics Group, Biological Sciences Division, Pacific Northwest National Laboratory, Richland, Washington 99352, United States
| | - Anthony J Cesnik
- Department of Genetics, Stanford University, Stanford, California 94305, United States
| | - Xinpei Yi
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas 77030, United States
| | - Xiaohu Hao
- Shanghai Institutes for Biological Science, Chinese Academy of Science, Shanghai 200031, China
| | - Michael R Shortreed
- Department of Chemistry, University of Wisconsin-Madison, Madison, Wisconsin 53706, United States
| | - Tujin Shi
- Integrative Omics Group, Biological Sciences Division, Pacific Northwest National Laboratory, Richland, Washington 99352, United States
| | - David M Lubman
- Department of Surgery, The University of Michigan, Ann Arbor, Michigan 48109, United States
| |
Collapse
|
20
|
Adams MR, Moody CT, Sollinger JL, Brudno Y. Extracellular-Matrix-Anchored Click Motifs for Specific Tissue Targeting. Mol Pharm 2020; 17:392-403. [PMID: 31829613 DOI: 10.1021/acs.molpharmaceut.9b00589] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Local presentation of cancer drugs by injectable drug-eluting depots reduces systemic side effects and improves efficacy. However, local depots deplete their drug stores and are difficult to introduce into stiff tissues, or organs, such as the brain, that cannot accommodate increased pressure. We present a method for introducing targetable depots through injection of activated ester molecules into target tissues that react with and anchor themselves to the local extracellular matrix (ECM) and subsequently capture systemically administered small molecules through bioorthogonal click chemistry. A computational model of tissue-anchoring depot formation and distribution was verified by histological analysis and confocal imaging of cleared tissues. ECM-anchored click groups do not elicit any noticeable local or systemic toxicity or immune response and specifically capture systemically circulating molecules at intradermal, intratumoral, and intracranial sites for multiple months. Taken together, ECM anchoring of click chemistry motifs is a promising approach to specific targeting of both small and large therapeutics, enabling repeated local presentation for cancer therapy and other diseases.
Collapse
Affiliation(s)
- Mary R Adams
- Joint Department of Biomedical Engineering , University of North Carolina, Chapel Hill and North Carolina State University , Raleigh. 911 Oval Drive , Raleigh , North Carolina 27695 , United States
| | - Christopher T Moody
- Joint Department of Biomedical Engineering , University of North Carolina, Chapel Hill and North Carolina State University , Raleigh. 911 Oval Drive , Raleigh , North Carolina 27695 , United States
| | - Jennifer L Sollinger
- Joint Department of Biomedical Engineering , University of North Carolina, Chapel Hill and North Carolina State University , Raleigh. 911 Oval Drive , Raleigh , North Carolina 27695 , United States
| | - Yevgeny Brudno
- Joint Department of Biomedical Engineering , University of North Carolina, Chapel Hill and North Carolina State University , Raleigh. 911 Oval Drive , Raleigh , North Carolina 27695 , United States.,Lineberger Comprehensive Cancer Center , University of North Carolina, Chapel Hill , 450 West Dr. , Chapel Hill , North Carolina 27599 , United States
| |
Collapse
|
21
|
Canon J, Rex K, Saiki AY, Mohr C, Cooke K, Bagal D, Gaida K, Holt T, Knutson CG, Koppada N, Lanman BA, Werner J, Rapaport AS, San Miguel T, Ortiz R, Osgood T, Sun JR, Zhu X, McCarter JD, Volak LP, Houk BE, Fakih MG, O'Neil BH, Price TJ, Falchook GS, Desai J, Kuo J, Govindan R, Hong DS, Ouyang W, Henary H, Arvedson T, Cee VJ, Lipford JR. The clinical KRAS(G12C) inhibitor AMG 510 drives anti-tumour immunity. Nature 2019; 575:217-223. [PMID: 31666701 DOI: 10.1038/s41586-019-1694-1] [Citation(s) in RCA: 1417] [Impact Index Per Article: 236.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Accepted: 09/18/2019] [Indexed: 12/24/2022]
Abstract
KRAS is the most frequently mutated oncogene in cancer and encodes a key signalling protein in tumours1,2. The KRAS(G12C) mutant has a cysteine residue that has been exploited to design covalent inhibitors that have promising preclinical activity3-5. Here we optimized a series of inhibitors, using novel binding interactions to markedly enhance their potency and selectivity. Our efforts have led to the discovery of AMG 510, which is, to our knowledge, the first KRAS(G12C) inhibitor in clinical development. In preclinical analyses, treatment with AMG 510 led to the regression of KRASG12C tumours and improved the anti-tumour efficacy of chemotherapy and targeted agents. In immune-competent mice, treatment with AMG 510 resulted in a pro-inflammatory tumour microenvironment and produced durable cures alone as well as in combination with immune-checkpoint inhibitors. Cured mice rejected the growth of isogenic KRASG12D tumours, which suggests adaptive immunity against shared antigens. Furthermore, in clinical trials, AMG 510 demonstrated anti-tumour activity in the first dosing cohorts and represents a potentially transformative therapy for patients for whom effective treatments are lacking.
Collapse
Affiliation(s)
- Jude Canon
- Amgen Research, Amgen Inc, Thousand Oaks, CA, USA.
| | - Karen Rex
- Amgen Research, Amgen Inc, Thousand Oaks, CA, USA
| | - Anne Y Saiki
- Amgen Research, Amgen Inc, Thousand Oaks, CA, USA
| | | | - Keegan Cooke
- Amgen Research, Amgen Inc, Thousand Oaks, CA, USA
| | | | - Kevin Gaida
- Amgen Research, Amgen Inc, Thousand Oaks, CA, USA
| | - Tyler Holt
- Amgen Research, Amgen Inc, Thousand Oaks, CA, USA
| | | | | | | | | | | | | | - Roberto Ortiz
- Amgen Research, Amgen Inc, Cambridge, MA, USA.,Pfizer, La Jolla, CA, USA
| | - Tao Osgood
- Amgen Research, Amgen Inc, Thousand Oaks, CA, USA
| | - Ji-Rong Sun
- Amgen Research, Amgen Inc, Thousand Oaks, CA, USA
| | - Xiaochun Zhu
- Amgen Research, Amgen Inc, Cambridge, MA, USA.,Takeda, Cambridge, MA, USA
| | | | - Laurie P Volak
- Amgen Research, Amgen Inc, Cambridge, MA, USA.,Celgene, San Diego, CA, USA
| | - Brett E Houk
- Amgen Clinical Development, Amgen Inc, Thousand Oaks, CA, USA
| | | | - Bert H O'Neil
- Indiana University School of Medicine, Indianapolis, IN, USA
| | - Timothy J Price
- The Queen Elizabeth Hospital, Woodville, South Australia, Australia.,University of Adelaide, Adelaide, South Australia, Australia
| | | | - Jayesh Desai
- Peter MacCallum Cancer Center, Melbourne, Victoria, Australia
| | - James Kuo
- Scientia Clinical Research, Randwick, New South Wales, Australia
| | | | - David S Hong
- The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Wenjun Ouyang
- Amgen Research, Amgen Inc, South San Francisco, CA, USA
| | - Haby Henary
- Amgen Clinical Development, Amgen Inc, Thousand Oaks, CA, USA
| | - Tara Arvedson
- Amgen Research, Amgen Inc, South San Francisco, CA, USA
| | - Victor J Cee
- Amgen Research, Amgen Inc, Thousand Oaks, CA, USA
| | | |
Collapse
|
22
|
Parakh S, King D, Gan HK, Scott AM. Current Development of Monoclonal Antibodies in Cancer Therapy. Recent Results Cancer Res 2019; 214:1-70. [PMID: 31473848 DOI: 10.1007/978-3-030-23765-3_1] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Exploiting the unique specificity of monoclonal antibodies has revolutionized the treatment and diagnosis of haematological and solid organ malignancies; bringing benefit to millions of patients over the past decades. Recent achievements include conjugating antibodies with toxic payloads resulting in superior efficacy and/or reduced toxicity, development of molecular imaging techniques targeting specific antigens for use as predictive and prognostic biomarkers, the development of novel bi- and tri-specific antibodies to enhance therapeutic benefit and abrogate resistance and the success of immunotherapy agents. In this chapter, we review an overview of antibody structure and function relevant to cancer therapy and provide an overview of pivotal clinical trials which have led to regulatory approval of monoclonal antibodies in cancer treatment. We further discuss resistance mechanisms and the unique side effects of each class of antibody and provide an overview of emerging therapeutic agents.
Collapse
Affiliation(s)
- Sagun Parakh
- Tumour Targeting Laboratory, Olivia Newton-John Cancer Research Institute, 145 Studley Road, Heidelberg, Melbourne, VIC, 3084, Australia.,Department of Medical Oncology, Olivia Newton-John Cancer and Wellness Centre, Austin Health, Heidelberg, Melbourne, Australia.,School of Cancer Medicine, La Trobe University, Melbourne, Australia
| | - Dylan King
- Tumour Targeting Laboratory, Olivia Newton-John Cancer Research Institute, 145 Studley Road, Heidelberg, Melbourne, VIC, 3084, Australia.,School of Cancer Medicine, La Trobe University, Melbourne, Australia
| | - Hui K Gan
- Tumour Targeting Laboratory, Olivia Newton-John Cancer Research Institute, 145 Studley Road, Heidelberg, Melbourne, VIC, 3084, Australia.,Department of Medical Oncology, Olivia Newton-John Cancer and Wellness Centre, Austin Health, Heidelberg, Melbourne, Australia.,School of Cancer Medicine, La Trobe University, Melbourne, Australia
| | - Andrew M Scott
- Tumour Targeting Laboratory, Olivia Newton-John Cancer Research Institute, 145 Studley Road, Heidelberg, Melbourne, VIC, 3084, Australia. .,School of Cancer Medicine, La Trobe University, Melbourne, Australia. .,Department of Molecular Imaging and Therapy, Austin Health, Melbourne, Australia. .,Department of Medicine, University of Melbourne, Melbourne, Australia.
| |
Collapse
|
23
|
Battaglin F, Puccini A, Ahcene Djaballah S, Lenz HJ. The impact of panitumumab treatment on survival and quality of life in patients with RAS wild-type metastatic colorectal cancer. Cancer Manag Res 2019; 11:5911-5924. [PMID: 31388315 PMCID: PMC6607986 DOI: 10.2147/cmar.s186042] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Accepted: 06/11/2019] [Indexed: 12/20/2022] Open
Abstract
Panitumumab is a fully human monoclonal antibody targeting the epidermal growth factor receptor (EGFR). It is currently approved for the treatment of RAS wild-type (WT) metastatic colorectal cancer (mCRC) in combination with chemotherapy in first- and second-line and as monotherapy in chemorefractory patients. This review will provide an overview of main efficacy data on panitumumab from its early development up to latest evidences, including novel perspectives on predictive biomarkers of anti-EGFRs efficacy and mechanisms of secondary resistance. Quality of life (QoL) related issues and panitumumab safety profile will be addressed as well.
Collapse
Affiliation(s)
- Francesca Battaglin
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Alberto Puccini
- Medical Oncology Unit 1, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Selma Ahcene Djaballah
- Medical Oncology Unit 1, Clinical and Experimental Oncology Department, Veneto Institute of Oncology IOV - IRCCS, Padua 35128, Italy
| | - Heinz-Josef Lenz
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| |
Collapse
|
24
|
A National Survey of Medical Oncologist's Opinions and Perceptions for Managing Rash Among mCRC Patients Treated with Panitumumab. Dermatol Ther (Heidelb) 2019; 9:337-353. [PMID: 31054146 PMCID: PMC6522609 DOI: 10.1007/s13555-019-0296-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Indexed: 10/27/2022] Open
Abstract
INTRODUCTION This study aimed to describe medical oncologist's opinions and perceptions regarding the management of dermatologic toxicities among metastatic colorectal cancer (mCRC) patients who were treated with panitumumab in the USA and assess if there were differences across demographic and clinical characteristics. METHODS We developed a survey based on the current literature and expert opinions regarding the management of dermatologic toxicities. The survey was implemented online in September 2016. Eligible oncologists were board certified and had treated at least five new or continuing patients with mCRC in the last 3 months, among whom at least three patients had received or were currently receiving panitumumab. RESULTS A total of 250 oncologists completed the survey. The data suggest that approximately 82% of patients received recommendations for moisturizer, 88% for sunscreen and 67% for ultraviolet (UV)-protective garments prior to or at the time of initiation of panitumumab therapy. There were minor differences in how dermatologic toxicities were managed across specific demographic or clinical groups. The data also suggest that the management associated with panitumumab use among mCRC patients can be greatly improved. CONCLUSIONS Our results highlight the urgent need for heightened education regarding dermatologic toxicity management among oncologists who treated mCRC patients with panitumumab. Easily implemented strategies, such as moisturizer, sunscreen, and UV-protective garments should be recommended to all patients. FUNDING Amgen, Inc. Plain language summary available for this article.
Collapse
|
25
|
Zou ZG, Rios FJ, Montezano AC, Touyz RM. TRPM7, Magnesium, and Signaling. Int J Mol Sci 2019; 20:E1877. [PMID: 30995736 PMCID: PMC6515203 DOI: 10.3390/ijms20081877] [Citation(s) in RCA: 94] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 04/12/2019] [Accepted: 04/12/2019] [Indexed: 12/17/2022] Open
Abstract
The transient receptor potential melastatin-subfamily member 7 (TRPM7) is a ubiquitously expressed chanzyme that possesses an ion channel permeable to the divalent cations Mg2+, Ca2+, and Zn2+, and an α-kinase that phosphorylates downstream substrates. TRPM7 and its homologue TRPM6 have been implicated in a variety of cellular functions and is critically associated with intracellular signaling, including receptor tyrosine kinase (RTK)-mediated pathways. Emerging evidence indicates that growth factors, such as EGF and VEGF, signal through their RTKs, which regulate activity of TRPM6 and TRPM7. TRPM6 is primarily an epithelial-associated channel, while TRPM7 is more ubiquitous. In this review we focus on TRPM7 and its association with growth factors, RTKs, and downstream kinase signaling. We also highlight how interplay between TRPM7, Mg2+ and signaling kinases influences cell function in physiological and pathological conditions, such as cancer and preeclampsia.
Collapse
Affiliation(s)
- Zhi-Guo Zou
- Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Centre, University of Glasgow, Glasgow G12 8TA, UK.
| | - Francisco J Rios
- Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Centre, University of Glasgow, Glasgow G12 8TA, UK.
| | - Augusto C Montezano
- Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Centre, University of Glasgow, Glasgow G12 8TA, UK.
| | - Rhian M Touyz
- Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Centre, University of Glasgow, Glasgow G12 8TA, UK.
| |
Collapse
|
26
|
Dote S, Itakura S, Kamei K, Hira D, Noda S, Kobayashi Y, Terada T. Oral mucositis associated with anti-EGFR therapy in colorectal cancer: single institutional retrospective cohort study. BMC Cancer 2018; 18:957. [PMID: 30290786 PMCID: PMC6173836 DOI: 10.1186/s12885-018-4862-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2017] [Accepted: 09/26/2018] [Indexed: 12/14/2022] Open
Abstract
Background Chemotherapy-induced oral mucositis impairs the quality of life. The difference in severity of oral mucositis between different anti-epidermal growth factor receptor (EGFR) antibodies combined with cytotoxic drugs in colorectal cancer is unclear. The aim of this study was to investigate the differences in oral mucositis between panitumumab (Pmab) and cetuximab (Cmab) combined with 5-fluorouracil (5-FU). Methods We conducted a retrospective cohort study. A total of 75 colorectal cancer outpatients treated with an anti-EGFR antibody combined with FOLFOX, FOLFIRI, or 5-FU/leucovorin as the first- to third-line treatment were included. The primary endpoint was the incidence of grade 2–3 oral mucositis. The secondary endpoint was the time to onset of oral mucositis. We also compared the incidence of toxicities of interest, skin toxicity, hypomagnesaemia and neutropenia, and time to treatment failure (TTF) between the two groups. Results Thirty-two patients treated with Pmab and 43 patients treated with Cmab were evaluated. Patient characteristics were similar between the two groups. The incidence of grade 2–3 oral mucositis was significantly higher with Pmab than with Cmab (31.3% vs 9.3%, P < 0.05). Moreover, the incidence of grade 3 oral mucositis was significantly higher in patients treated with Pmab (18.8% vs 0%, P < 0.01). The mean (SD) cycles to onset of the worst oral mucositis was 3.0 (2.9) in the Pmab group and 2.3 (1.7) in the Cmab group (P = 0.29). Oral mucositis was characterized by glossitis and cheilitis. The incidences of other toxicities were the following (Pmab vs Cmab): grade 2–3 skin toxicity: 68.8% vs 74.4% (P = 0.61), grade 2–3 hypomagnesaemia: 9.3% vs 7.0% (P = 1.00), grade 3–4 neutropenia: 28.1% vs 37.2% (P = 0.46). The median TTF was not significantly different, i.e., 223 days vs 200 days (P = 0.39) for Pmab vs Cmab. Conclusions Pmab-based chemotherapy resulted in significantly higher grades of oral mucositis compared with Cmab-based chemotherapy. The oral condition should be monitored carefully and early supportive care should be provided for patients treated with Pmab-based chemotherapy. Electronic supplementary material The online version of this article (10.1186/s12885-018-4862-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Satoshi Dote
- Department of Pharmacy, Kyoto-Katsura Hospital, 17, Yamadahiraocho, Kyoto-shi Nishikyo-ku, Kyoto, 615-8256, Japan. .,Department of Pharmacy, Shiga University of Medical Science Hospital, Seta Tsukinowa-cho, Otsu, Shiga, 520-2192, Japan.
| | - Shoji Itakura
- Department of Pharmacy, Japanese Red Cross Kyoto Daiichi Hospital, 15-749, Hommachi, Kyoto-shi Higashiyama-ku, Kyoto, 605-0981, Japan
| | - Kohei Kamei
- Department of Pharmacy, Kyoto-Katsura Hospital, 17, Yamadahiraocho, Kyoto-shi Nishikyo-ku, Kyoto, 615-8256, Japan
| | - Daiki Hira
- Department of Pharmacy, Shiga University of Medical Science Hospital, Seta Tsukinowa-cho, Otsu, Shiga, 520-2192, Japan.,College of Pharmaceutical Sciences, Ritsumeikan University, Noji-higashi 1-1-1, Kusatsu, Shiga, 525-8577, Japan
| | - Satoshi Noda
- Department of Pharmacy, Shiga University of Medical Science Hospital, Seta Tsukinowa-cho, Otsu, Shiga, 520-2192, Japan
| | - Yuka Kobayashi
- Department of Pharmacy, Kyoto-Katsura Hospital, 17, Yamadahiraocho, Kyoto-shi Nishikyo-ku, Kyoto, 615-8256, Japan
| | - Tomohiro Terada
- Department of Pharmacy, Shiga University of Medical Science Hospital, Seta Tsukinowa-cho, Otsu, Shiga, 520-2192, Japan
| |
Collapse
|
27
|
Mao C, Zhao Y, Li F, Li Z, Tian S, Debinski W, Ming X. P-glycoprotein targeted and near-infrared light-guided depletion of chemoresistant tumors. J Control Release 2018; 286:289-300. [PMID: 30081143 DOI: 10.1016/j.jconrel.2018.08.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2018] [Revised: 07/31/2018] [Accepted: 08/02/2018] [Indexed: 01/07/2023]
Abstract
Drug resistance remains a formidable challenge to cancer therapy. P-glycoprotein (Pgp) contributes to multidrug resistance in numerous cancers by preventing accumulation of anticancer drugs in cancer cells. Strategies to overcome this resistance have been vigorously sought for over 3 decades, yet clinical solutions do not exist. The main reason for the failure is lack of cancer specificity of small-molecule Pgp inhibitors, thus causing severe toxicity in normal tissues. In this study, Pgp-targeted photodynamic therapy (PDT) was developed to achieve superior cancer specificity through antibody targeting plus locoregional light activation. Thus, a Pgp monoclonal antibody was chemically modified with IR700, a porphyrin photosensitizer. In vitro studies showed that the antibody-photosensitizer conjugates specifically bind to Pgp-expressing drug resistant cancer cells, and caused dramatic cytotoxicity upon irradiation with a near infrared light. We then tested our Pgp-targeted approach in mouse xenograft models of chemoresistant ovarian cancer and head and neck cancer. In both models, targeted PDT produced rapid tumor shrinkage, and significantly prolonged survival of tumor-bearing mice. We conclude that our targeted PDT approach produces molecularly targeted and spatially selective ablation of chemoresistant tumors, and thereby provides an effective approach to overcome Pgp-mediated multidrug resistance in cancer, where conventional approaches have failed.
Collapse
Affiliation(s)
- Chengqiong Mao
- Departments of Cancer Biology and Biomedical Engineering, Comprehensive Cancer Center, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | - Yan Zhao
- Departments of Cancer Biology and Biomedical Engineering, Comprehensive Cancer Center, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | - Fang Li
- Departments of Cancer Biology and Biomedical Engineering, Comprehensive Cancer Center, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | - Zibo Li
- Department of Radiology and Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Shaomin Tian
- Department of Microbiology & Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Waldemar Debinski
- Departments of Cancer Biology and Biomedical Engineering, Comprehensive Cancer Center, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA; Brain Tumor Center of Excellence, Thomas K Hearn Brain Tumor Research Center, Winston-Salem, NC 27157, USA
| | - Xin Ming
- Departments of Cancer Biology and Biomedical Engineering, Comprehensive Cancer Center, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA.
| |
Collapse
|
28
|
Angeles A, Hung W, Cheung WY. Eligibility of real-world patients with chemo-refractory, K-RAS wild-type, metastatic colorectal cancer for palliative intent regorafenib monotherapy. Med Oncol 2018; 35:114. [PMID: 29936654 DOI: 10.1007/s12032-018-1176-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Accepted: 06/20/2018] [Indexed: 12/11/2022]
Abstract
The CORRECT trial demonstrated survival benefits with regorafenib monotherapy in patients with treatment-refractory, metastatic colorectal cancer (mCRC). However, the trial's stringent eligibility criteria for regorafenib may limit its external validity. We aimed to examine treatment attrition rates and eligibility for regorafenib in routine practice. We identified patients at the British Columbia Cancer Agency diagnosed with mCRC who demonstrated disease progression or intolerable toxicity on 2 or more lines of systemic therapy. During the study timeframe, panitumumab and cetuximab were only used in the chemo-refractory setting. Data on clinicopathologic variables and patient outcomes were ascertained and analyzed. Eligibility was determined using the CORRECT trial criteria. A total of 391 patients were identified, among whom only 39% were eligible for regorafenib: 35% in the panitumumab group and 51% in the cetuximab group. The main reasons for ineligibility in all patients were Eastern Cooperative Oncology Group Performance Status (ECOG PS) > 1 (69%), an elevated total bilirubin (21%), and thromboembolic events in the past 6 months (10%). No difference in eligibility for regorafenib was observed between patients previously receiving panitumumab or cetuximab (P = 0.914; 95% CI 0.550-1.951). Kaplan-Meier analyses showed that regorafenib-eligible compared to regorafenib-ineligible patients had an increased median overall survival of 5.3 versus 2.1 months, respectively (P < 0.001). However, Cox proportional hazard analyses showed that only ECOG PS rather than trial eligibility was correlated with outcomes. The strict eligibility criteria disqualify most patients with treatment-refractory mCRC for regorafenib therapy. Future trials should broaden the eligibility criteria to improve external validity.
Collapse
Affiliation(s)
- Arkhjamil Angeles
- Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Wayne Hung
- Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Winson Y Cheung
- Department of Oncology, Tom Baker Cancer Center, University of Calgary, 1331 29 Street NW, Calgary, AB, T2N4N2, Canada.
- Health Services Research, Cancer Control Alberta, Calgary, AB, Canada.
- Cancer Health Outcomes Research Database (CHORD) Consortium, Calgary, AB, Canada.
| |
Collapse
|
29
|
Shin W, Lee SK, Hwang JH, Park JC, Cho YH, Ro EJ, Song Y, Seo HR, Choi KY. Identification of Ras-degrading small molecules that inhibit the transformation of colorectal cancer cells independent of β-catenin signaling. Exp Mol Med 2018; 50:1-10. [PMID: 29884842 PMCID: PMC5994827 DOI: 10.1038/s12276-018-0102-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Accepted: 03/06/2018] [Indexed: 12/17/2022] Open
Abstract
Although the development of drugs that control Ras is an emerging topic in cancer therapy, no clinically applicable drug is currently available. We have previously utilized knowledge of the Wnt/β-catenin signaling-dependent mechanism of Ras protein stability regulation to identify small molecules that inhibit the proliferation and transformation of various colorectal cancer (CRC) cells via degradation of both β-catenin and Ras. Due to the absence of Ras degradation in cells expressing a nondegradable mutant form of β-catenin and the need to determine an alternative mechanism of Ras degradation, we designed a cell-based system to screen compounds that degrade Ras independent of the Wnt/β-catenin signaling pathway. A cell-based high-content screening (HCS) system that monitors the levels of EGFP-K-RasG12V was established using HCT-116 cells harboring a nondegradable mutant CTNNB1 (ΔS45). Through HCS of a chemical library composed of 10,000 compounds and subsequent characterization of hits, we identified several compounds that degrade Ras without affecting the β-catenin levels. KY7749, one of the most effective compounds, inhibited the proliferation and transformation of CRC cells, especially KRAS-mutant cells that are resistant to the EGFR monoclonal antibody cetuximab. Small molecules that degrade Ras independent of β-catenin may able to be used in treatments for cancers caused by aberrant EGFR and Ras. Mutations in KRAS, a gene regulating cell proliferation, occur in 40–50% of colorectal cancer (CRC) patients. These cancers are usually insensitive to antibody drugs targeting the epidermal growth factor receptor (EGFR) on cancer cells. Kang-Yell Choi at Yonsei University, Seoul, South Korea and co-workers had previously identified small molecules that inhibited CRC by degrading the signaling proteins β-catenin and Ras. However, CRC cells with β-catenin mutations are resistant to Ras-degrading compounds. In this study, they screened 10,000 small molecules that could degrade Ras in CRC cells expressing nondegradable mutant β-catenin. They identified small molecules that inhibited growth of CRC cells resistant to EGFR-mediated antibody therapy. Further investigation of these molecules may help develop new drugs for treating CRC.
Collapse
Affiliation(s)
- Wookjin Shin
- Translational Research Center for Protein Function Control, Yonsei University, Seoul, Republic of Korea.,Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Sang-Kyu Lee
- Translational Research Center for Protein Function Control, Yonsei University, Seoul, Republic of Korea.,Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Jeong-Ha Hwang
- Translational Research Center for Protein Function Control, Yonsei University, Seoul, Republic of Korea.,Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Jong-Chan Park
- Translational Research Center for Protein Function Control, Yonsei University, Seoul, Republic of Korea.,Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Yong-Hee Cho
- Translational Research Center for Protein Function Control, Yonsei University, Seoul, Republic of Korea.,Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Eun Ji Ro
- Translational Research Center for Protein Function Control, Yonsei University, Seoul, Republic of Korea.,Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Yeonhwa Song
- Cancer Biology Research Laboratory, Institut Pasteur Korea, Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Haeng Ran Seo
- Cancer Biology Research Laboratory, Institut Pasteur Korea, Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Kang-Yell Choi
- Translational Research Center for Protein Function Control, Yonsei University, Seoul, Republic of Korea. .,Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea.
| |
Collapse
|
30
|
Fornasier G, Taborelli M, Francescon S, Polesel J, Aliberti M, De Paoli P, Baldo P. Targeted therapies and adverse drug reactions in oncology: the role of clinical pharmacist in pharmacovigilance. Int J Clin Pharm 2018; 40:795-802. [PMID: 29785683 PMCID: PMC6132980 DOI: 10.1007/s11096-018-0653-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Accepted: 05/08/2018] [Indexed: 12/30/2022]
Abstract
Background The majority of adverse drug reactions (ADRs) reported in the summary of product characteristics (SPCs) are based on pivotal clinical trials, performed under controlled conditions and with selected patients. Objectives (1) to observe ADRs in the real-world setting and to evaluate if the supervision of the pharmacist impacts on the management of ADRs and on the satisfaction of patients; (2) to sensitise health professionals and patients on the need to increase the reporting of ADRs, in compliance with Pharmacovigilance. Setting CRO Aviano, Italian National Cancer Institute. Method From February 2013 to April 2015, we conducted an observational study enrolling 154 patients (≥ 18 years) undergoing treatment with at least one of ten targeted-therapies included in the study. Main outcome ADR reporting in the real-world setting. Patient satisfaction with clinical pharmacist support. Results Reported ADRs in the real setting do not always correspond with data described in the respective SPCs. Unknown ADRs were also identified such as hyperglycaemia with lenalidomide and sorafenib; and hypomagnesaemia with bevacizumab. We also observed a 124.3% increase in spontaneous reports. Conclusion This study shows the high value of active pharmacovigilance programs, and our results might be a starting point for developing a randomised trial which should aim to demonstrate the impact of the pharmacist on improving patient’s adherence and in measuring the difference in ADRs reports in the different arms followed or not by the pharmacist.
Collapse
Affiliation(s)
- G Fornasier
- Pharmacy Unit, CRO Aviano National Cancer Institute - IRCCS, Via F. Gallini 2, 33080, Aviano, PN, Italy
| | - M Taborelli
- Unit of Cancer Epidemiology, CRO Aviano National Cancer Institute - IRCCS, Via F. Gallini 2, 33080, Aviano, Italy
| | - S Francescon
- Pharmacy Unit, CRO Aviano National Cancer Institute - IRCCS, Via F. Gallini 2, 33080, Aviano, PN, Italy
| | - J Polesel
- Unit of Cancer Epidemiology, CRO Aviano National Cancer Institute - IRCCS, Via F. Gallini 2, 33080, Aviano, Italy
| | - M Aliberti
- Pharmacy Unit, CRO Aviano National Cancer Institute - IRCCS, Via F. Gallini 2, 33080, Aviano, PN, Italy
| | - P De Paoli
- Scientific Directorate, CRO Aviano National Cancer Institute - IRCCS, Via F. Gallini 2, 33080, Aviano, PN, Italy
| | - P Baldo
- Pharmacy Unit, CRO Aviano National Cancer Institute - IRCCS, Via F. Gallini 2, 33080, Aviano, PN, Italy.
| |
Collapse
|
31
|
Goss GD, Vokes EE, Gordon MS, Gandhi L, Papadopoulos KP, Rasco DW, Fischer JS, Chu KL, Ames WW, Mittapalli RK, Lee HJ, Zeng J, Roberts-Rapp LA, Loberg LI, Ansell PJ, Reilly EB, Ocampo CJ, Holen KD, Tolcher AW. Efficacy and safety results of depatuxizumab mafodotin (ABT-414) in patients with advanced solid tumors likely to overexpress epidermal growth factor receptor. Cancer 2018. [PMID: 29533458 PMCID: PMC5969257 DOI: 10.1002/cncr.31304] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
BACKGROUND Epidermal growth factor receptor (EGFR) alterations are associated with multiple cancers. Current EGFR-directed therapies have led to increased efficacy but are associated with specific side effects. The antibody-drug conjugate depatuxizumab mafodotin (depatux-m) targets EGFR with a monoclonal antibody linked to a cytotoxin, and is highly tumor-specific. METHODS This phase 1/2 study evaluated the safety, pharmacokinetics, and efficacy of depatux-m in patients who had advanced solid tumors with known wild-type EGFR overexpression, amplification, or mutated EGFR variant III. A 3 + 3 dose escalation was used, and 2 dosing schedules were evaluated. Depatux-m also was manufactured under an alternate process to reduce the drug load and improve the safety profile, and it was tested at the maximum tolerated dose (MTD). In another cohort, prolonged infusion time of depatux-m was evaluated; and a cohort with confirmed EGFR amplification also was evaluated at the MTD. RESULTS Fifty-six patients were treated. The MTD and the recommended phase 2 dose for depatux-m was 3.0 mg/kg. Common adverse events (AEs) were blurred vision (48%) and fatigue (41%). A majority of patients (66%) experienced 1 or more ocular AEs. Grade 3 or 4 AEs were observed in 43% of patients. One patient with EGFR-amplified, triple-negative breast cancer had a partial response. Stable disease was observed in 23% of patients. Pharmacokinetics revealed that depatux-m exposures were approximately dose-proportional. CONCLUSIONS Depatux-m resulted in infrequent nonocular AEs but increased ocular AEs. Patient follow-up confirmed that ocular AEs were reversible. Lowering the drug-antibody ratio did not decrease the number of ocular AEs. A partial response in 1 patient with EGFR-amplified disease provides the opportunity to study depatux-m in diseases with a high incidence of EGFR amplification. Cancer 2018;124:2174-83. © 2018 The Authors. Cancer published by Wiley Periodicals, Inc. on behalf of American Cancer Society. This is an open access article under the terms of the Creative Commons Attribution NonCommercial License, which permits use, distribution and reproduction in any medium, provided the original work is properly cited and is not used for commercial purposes.
Collapse
Affiliation(s)
- Glenwood D Goss
- The Ottawa Hospital Research Institute and the University of Ottawa, Ottawa, Ontario, Canada
| | - Everett E Vokes
- Department of Medicine, University of Chicago, Chicago, Illinois
| | | | - Leena Gandhi
- New York University Perlmutter Cancer Center, NYU Health, New York, New York
| | | | - Drew W Rasco
- South Texas Accelerated Research Therapeutics, San Antonio, Texas
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Bhullar KS, Lagarón NO, McGowan EM, Parmar I, Jha A, Hubbard BP, Rupasinghe HPV. Kinase-targeted cancer therapies: progress, challenges and future directions. Mol Cancer 2018; 17:48. [PMID: 29455673 PMCID: PMC5817855 DOI: 10.1186/s12943-018-0804-2] [Citation(s) in RCA: 790] [Impact Index Per Article: 112.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2017] [Accepted: 02/01/2018] [Indexed: 02/06/2023] Open
Abstract
The human genome encodes 538 protein kinases that transfer a γ-phosphate group from ATP to serine, threonine, or tyrosine residues. Many of these kinases are associated with human cancer initiation and progression. The recent development of small-molecule kinase inhibitors for the treatment of diverse types of cancer has proven successful in clinical therapy. Significantly, protein kinases are the second most targeted group of drug targets, after the G-protein-coupled receptors. Since the development of the first protein kinase inhibitor, in the early 1980s, 37 kinase inhibitors have received FDA approval for treatment of malignancies such as breast and lung cancer. Furthermore, about 150 kinase-targeted drugs are in clinical phase trials, and many kinase-specific inhibitors are in the preclinical stage of drug development. Nevertheless, many factors confound the clinical efficacy of these molecules. Specific tumor genetics, tumor microenvironment, drug resistance, and pharmacogenomics determine how useful a compound will be in the treatment of a given cancer. This review provides an overview of kinase-targeted drug discovery and development in relation to oncology and highlights the challenges and future potential for kinase-targeted cancer therapies.
Collapse
Affiliation(s)
- Khushwant S Bhullar
- Department of Pharmacology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Naiara Orrego Lagarón
- Department of Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, University of Barcelona, Barcelona, Spain
| | - Eileen M McGowan
- Chronic Disease Solutions Team, School of Life Science, University of Technology, New South Wales, Australia
| | - Indu Parmar
- Division of Product Development, Radient Technologies, Edmonton, AB, Canada
| | - Amitabh Jha
- Department of Chemistry, Acadia University, Wolfville, NS, Canada
| | - Basil P Hubbard
- Department of Pharmacology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - H P Vasantha Rupasinghe
- Department of Plant, Food, and Environmental Sciences, Faculty of Agriculture, Dalhousie University, Truro, NS, Canada.
- Department of Pathology, Faculty of Medicine, Dalhousie University, Halifax, NS, Canada.
| |
Collapse
|
33
|
Dermatologic Toxicity Occurring During Anti-EGFR Monoclonal Inhibitor Therapy in Patients With Metastatic Colorectal Cancer: A Systematic Review. Clin Colorectal Cancer 2017; 17:85-96. [PMID: 29576427 PMCID: PMC6773267 DOI: 10.1016/j.clcc.2017.12.004] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Revised: 11/13/2017] [Accepted: 12/07/2017] [Indexed: 12/22/2022]
Abstract
Monoclonal antibody inhibitors of the epidermal growth factor receptor (EGFR) have been shown to improve outcomes for patients with metastatic colorectal cancer (mCRC) without RAS gene mutations. However, treatment with anti-EGFR agents can be associated with toxicities of the skin, nails, hair, and eyes. Because these dermatologic toxicities can result in treatment discontinuation and affect patient quality of life, their management is an important focus when administering anti-EGFR monoclonal antibodies. The present systematic review describes the current data reporting the nature and incidence of, and management and treatment options for, dermatologic toxicities occurring during anti-EGFR treatment of mCRC. A search of the National Library of Medicine PubMed database from January 1, 2009, to August 18, 2016, identified relevant reports discussing dermatologic toxicity management among patients with mCRC receiving anti-EGFR therapy. The studies were grouped by type and rated by level of evidence using the GRADE approach developed by the Agency for Healthcare Research and Quality. Overall, 269 reports were reviewed (nonrandomized trials, n = 120; randomized trials, n = 31; retrospective studies, n = 15; reviews, n = 39). Dermatologic toxicity of any grade occurs in most patients who receive anti-EGFR therapy; approximately 10% to 20% of patients experienced grade 3/4 toxicity. The most common dermatologic toxicities include papulopustular/acneiform rash, xerosis, and pruritus; however, nail changes, hair abnormalities, and ocular conditions also occur. Guidance for managing these toxicities includes the use of inexpensive emollient ointments and moisturizers, avoidance of sun exposure, avoidance of irritants, and the use of short showers. Several studies also found that preemptive treatment was more effective than reactive treatment at limiting the incidence and severity of skin toxicity. With appropriate treatment, the dermatologic toxicities associated with anti-EGFR monoclonal antibody therapy can be managed, minimizing patient discomfort and the need for therapy interruption and/or discontinuation. Additionally, preemptive treatment can reduce dermatologic toxicity severity, ultimately yielding better quality of life.
Collapse
|
34
|
van den Bent M, Gan HK, Lassman AB, Kumthekar P, Merrell R, Butowski N, Lwin Z, Mikkelsen T, Nabors LB, Papadopoulos KP, Penas-Prado M, Simes J, Wheeler H, Walbert T, Scott AM, Gomez E, Lee HJ, Roberts-Rapp L, Xiong H, Bain E, Ansell PJ, Holen KD, Maag D, Reardon DA. Efficacy of depatuxizumab mafodotin (ABT-414) monotherapy in patients with EGFR-amplified, recurrent glioblastoma: results from a multi-center, international study. Cancer Chemother Pharmacol 2017; 80:1209-1217. [PMID: 29075855 PMCID: PMC5686264 DOI: 10.1007/s00280-017-3451-1] [Citation(s) in RCA: 93] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Accepted: 10/07/2017] [Indexed: 11/06/2022]
Abstract
PURPOSE Patients with recurrent glioblastoma (rGBM) have a poor prognosis. Epidermal growth factor receptor (EGFR) gene amplification is present in ~ 50% of glioblastomas (GBMs). Depatuxizumab mafodotin (depatux-m), formerly ABT-414, is an antibody-drug conjugate that preferentially binds cells with EGFR amplification, is internalized and releases a potent antimicrotubule agent, monomethyl auristatin F (MMAF). Here we report the safety, pharmacokinetics, and efficacy of depatux-m monotherapy at the recommended Phase 2 dose (RPTD) in patients with EGFR-amplified, rGBM. METHODS M12-356 (NCT01800695) is an open-label study with three escalation and expansion cohorts. Sixty-six patients with EGFR-amplified, rGBM were treated with depatux-m monotherapy at 1.25 mg/kg intravenously every 2 weeks. Adults with measurable rGBM, who were bevacizumab-naïve, with EGFR amplification were eligible. RESULTS Among 66 patients, median age was 58 years (range 35-80). All patients were previously treated with radiotherapy/temozolomide. The most common adverse events (AEs) were eye related (91%), including blurred vision (65%), dry eye (29%), keratitis, and photophobia (27% each). Grade 3/4 AEs occurred in 42% of all patients, and ocular Grade 3/4 AEs occurred in 33% of patients overall. One patient (2%) had a Grade 4 ocular AE. Ocular AEs were manageable and usually resolved once treatment with depatux-m ceased. The objective response rate was 6.8%, the 6-month progression-free survival rate was 28.8%, and the 6-month overall survival rate was 72.5%. CONCLUSION Depatux-m monotherapy displayed frequent but mostly Grade 1/2 ocular toxicities. A PFS6 of 28.8% was observed in this rGBM population, warranting further study.
Collapse
Affiliation(s)
- Martin van den Bent
- Brain Tumor Center, Erasmus MC Cancer Institute, Groene Hilledijk 301, 3075 EA, Rotterdam, the Netherlands.
| | - Hui K Gan
- School of Cancer Medicine, La Trobe University, Melbourne, VIC, Australia
- Department of Medicine, University of Melbourne, Melbourne, VIC, Australia
| | - Andrew B Lassman
- Department of Neurology and Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, NY, USA
| | | | - Ryan Merrell
- NorthShore University Health System, Evanston, IL, USA
| | - Nicholas Butowski
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Zarnie Lwin
- Department of Medical Oncology, School of Medicine, University of Queensland, Royal Brisbane and Women's Hospital, Brisbane, Australia
| | | | - Louis B Nabors
- University of Alabama at Birmingham, Birmingham, AL, USA
| | | | | | - John Simes
- NHMRC Clinical Trials Centre, University of Sydney, Sydney, NSW, Australia
| | - Helen Wheeler
- Medical Oncology, Royal North Shore Hospital, Sydney, NSW, Australia
| | | | - Andrew M Scott
- School of Cancer Medicine, La Trobe University, Melbourne, VIC, Australia
- Department of Medicine, University of Melbourne, Melbourne, VIC, Australia
| | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
de Bruin RCG, Veluchamy JP, Lougheed SM, Schneiders FL, Lopez-Lastra S, Lameris R, Stam AG, Sebestyen Z, Kuball J, Molthoff CFM, Hooijberg E, Roovers RC, Santo JPD, van Bergen En Henegouwen PMP, Verheul HMW, de Gruijl TD, van der Vliet HJ. A bispecific nanobody approach to leverage the potent and widely applicable tumor cytolytic capacity of Vγ9Vδ2-T cells. Oncoimmunology 2017; 7:e1375641. [PMID: 29296532 DOI: 10.1080/2162402x.2017.1375641] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Revised: 08/11/2017] [Accepted: 08/31/2017] [Indexed: 12/23/2022] Open
Abstract
Though Vγ9Vδ2-T cells constitute only a small fraction of the total T cell population in human peripheral blood, they play a vital role in tumor defense and are therefore of major interest to explore for cancer immunotherapy. Vγ9Vδ2-T cell-based cancer immunotherapeutic approaches developed so far have been generally well tolerated and were able to induce significant clinical responses. However, overall results were inconsistent, possibly due to the fact that these strategies induced systemic activation of Vγ9Vδ2-T cells without preferential accumulation and targeted activation in the tumor. Here we show that a novel bispecific nanobody-based construct targeting both Vγ9Vδ2-T cells and EGFR induced potent Vγ9Vδ2-T cell activation and subsequent tumor cell lysis both in vitro and in an in vivo mouse xenograft model. Tumor cell lysis was independent of KRAS and BRAF tumor mutation status and common Vγ9Vδ2-T cell receptor sequence variations. In combination with the conserved monomorphic nature of the Vγ9Vδ2-TCR and the facile replacement of the tumor-specific nanobody, this immunotherapeutic approach can be applied to a large group of cancer patients.
Collapse
Affiliation(s)
- Renée C G de Bruin
- Department of Medical Oncology, VU University Medical Center, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - John P Veluchamy
- Department of Medical Oncology, VU University Medical Center, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - Sinéad M Lougheed
- Department of Medical Oncology, VU University Medical Center, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - Famke L Schneiders
- Department of Medical Oncology, VU University Medical Center, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - Silvia Lopez-Lastra
- Innate Immunity Unit, Institut Pasteur, Paris, France.,Institut National de la Santé et de la Recherche Médicale (INSERM) U1223, Paris, France.,Université Paris-Sud, Université Paris-Saclay, Gif-sur-Yvette, France
| | - Roeland Lameris
- Department of Medical Oncology, VU University Medical Center, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - Anita G Stam
- Department of Medical Oncology, VU University Medical Center, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - Zsolt Sebestyen
- Department of Hematology and Laboratory of Translational Immunology, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands
| | - Jürgen Kuball
- Department of Hematology and Laboratory of Translational Immunology, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands
| | - Carla F M Molthoff
- Department of Radiology and Nuclear Medicine, VU University Medical Center, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - Erik Hooijberg
- Department of Pathology, VU University Medical Center, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - Rob C Roovers
- Department of Cell Biology, Faculty of Science, Utrecht University, Padualaan 8, 3584 CH, Utrecht, The Netherlands
| | - James P Di Santo
- Innate Immunity Unit, Institut Pasteur, Paris, France.,Institut National de la Santé et de la Recherche Médicale (INSERM) U1223, Paris, France
| | | | - Henk M W Verheul
- Department of Medical Oncology, VU University Medical Center, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - Tanja D de Gruijl
- Department of Medical Oncology, VU University Medical Center, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - Hans J van der Vliet
- Department of Medical Oncology, VU University Medical Center, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| |
Collapse
|
36
|
Scavone C, Sportiello L, Sullo MG, Ferrajolo C, Ruggiero R, Sessa M, Berrino PM, di Mauro G, Berrino L, Rossi F, Rafaniello C, Capuano A. Safety Profile of Anticancer and Immune-Modulating Biotech Drugs Used in a Real World Setting in Campania Region (Italy): BIO-Cam Observational Study. Front Pharmacol 2017; 8:607. [PMID: 28932193 PMCID: PMC5592230 DOI: 10.3389/fphar.2017.00607] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Accepted: 08/22/2017] [Indexed: 12/19/2022] Open
Abstract
Objectives: To investigate the occurrence of adverse events (AEs) in naïve patients receiving biotech drugs. Design: A prospective observational study. Setting: Onco-hematology, Hepato-gastroenterology, Rheumatology, Dermatology, and Neurology Units in Campania Region (Italy). Participants: 775 patients (53.81% female) with mean age 56.0 (SD 15.2). The mean follow-up/patient was 3.48 (95% confidence interval 3.13–3.84). Main outcome measures: We collected all AEs associated to biotech drugs, including serious infections and malignancies. Serious AEs were defined according to the International Conference on Harmonization of Technical Requirements for Registration of Pharmaceuticals for Human Use, clinical safety data management: definitions and standards for expedited reporting E2A guideline. Results: The majority of the study population was enrolled in Onco-hematology and Rheumatology Units and the most common diagnosis were hematological malignancies, followed by rheumatoid arthritis, colorectal cancer, breast cancer, and psoriatic arthritis. The most commonly prescribed biotech drugs were rituximab, bevacizumab, infliximab, trastuzumab, adalimumab, and cetuximab. Out of 775 patients, 320 experienced at least one AE. Most of patients experienced AEs to cetuximab therapy, rituximab and trastuzumab. Comparing female and male population, our findings highlighted a statistically significant difference in terms of AEs for adalimumab (35.90% vs. 7.41%, p < 0.001) and etanercept (27.59% vs. 10.00%, p = 0.023). Considering all biotech drugs, we observed a peak for all AEs occurrence at follow-up 91–180 days category. Bevacizumab, brentuximab, rituximab, trastuzumab and cetuximab were more commonly associated to serious adverse events; most of these were possibly related to biotech drugs, according to causality assessment. Three cases of serious infections occurred. Conclusions: The results of our study demonstrated that the majority of AEs were not serious and expected. Few cases of serious infections occurred, while no case of malignancy did. Overall, the safety profile of biotech drugs used in our population was similar to those observed in pivotal trials. Notwithstanding the positive results of our study, some safety concerns still remain unresolved. In order to collect more effectiveness and safety data on biotech drugs, the collection and analysis of real world data should be endorsed as well as the management of post-authorization studies.
Collapse
Affiliation(s)
- Cristina Scavone
- The authors would like to thank all the members of the BIO-Cam group who provided patient data for this study: University Hospital of Università degli Studi della Campania "Luigi Vanvitelli" Naples; Hospital SG Moscati-Avellino; Istituto Nazionale Tumori-IRCCS "Fondazione G. Pascale" Naples; Hospital AORN Cardarelli Naples; Hospital G Rummo Benevento; Hospital Sant'Anna e San Sebastiano Caserta; University Hospital Università degli Studi di Napoli Federico II Naples; Fondazione Maugeri Benevento; University Hospital San Giovanni di Dio e Ruggi d'Aragona Salerno; Hospital Ospedale dei Colli Naples
| | - Liberata Sportiello
- The authors would like to thank all the members of the BIO-Cam group who provided patient data for this study: University Hospital of Università degli Studi della Campania "Luigi Vanvitelli" Naples; Hospital SG Moscati-Avellino; Istituto Nazionale Tumori-IRCCS "Fondazione G. Pascale" Naples; Hospital AORN Cardarelli Naples; Hospital G Rummo Benevento; Hospital Sant'Anna e San Sebastiano Caserta; University Hospital Università degli Studi di Napoli Federico II Naples; Fondazione Maugeri Benevento; University Hospital San Giovanni di Dio e Ruggi d'Aragona Salerno; Hospital Ospedale dei Colli Naples
| | - Maria G Sullo
- The authors would like to thank all the members of the BIO-Cam group who provided patient data for this study: University Hospital of Università degli Studi della Campania "Luigi Vanvitelli" Naples; Hospital SG Moscati-Avellino; Istituto Nazionale Tumori-IRCCS "Fondazione G. Pascale" Naples; Hospital AORN Cardarelli Naples; Hospital G Rummo Benevento; Hospital Sant'Anna e San Sebastiano Caserta; University Hospital Università degli Studi di Napoli Federico II Naples; Fondazione Maugeri Benevento; University Hospital San Giovanni di Dio e Ruggi d'Aragona Salerno; Hospital Ospedale dei Colli Naples
| | - Carmen Ferrajolo
- The authors would like to thank all the members of the BIO-Cam group who provided patient data for this study: University Hospital of Università degli Studi della Campania "Luigi Vanvitelli" Naples; Hospital SG Moscati-Avellino; Istituto Nazionale Tumori-IRCCS "Fondazione G. Pascale" Naples; Hospital AORN Cardarelli Naples; Hospital G Rummo Benevento; Hospital Sant'Anna e San Sebastiano Caserta; University Hospital Università degli Studi di Napoli Federico II Naples; Fondazione Maugeri Benevento; University Hospital San Giovanni di Dio e Ruggi d'Aragona Salerno; Hospital Ospedale dei Colli Naples
| | - Rosanna Ruggiero
- The authors would like to thank all the members of the BIO-Cam group who provided patient data for this study: University Hospital of Università degli Studi della Campania "Luigi Vanvitelli" Naples; Hospital SG Moscati-Avellino; Istituto Nazionale Tumori-IRCCS "Fondazione G. Pascale" Naples; Hospital AORN Cardarelli Naples; Hospital G Rummo Benevento; Hospital Sant'Anna e San Sebastiano Caserta; University Hospital Università degli Studi di Napoli Federico II Naples; Fondazione Maugeri Benevento; University Hospital San Giovanni di Dio e Ruggi d'Aragona Salerno; Hospital Ospedale dei Colli Naples
| | - Maurizio Sessa
- The authors would like to thank all the members of the BIO-Cam group who provided patient data for this study: University Hospital of Università degli Studi della Campania "Luigi Vanvitelli" Naples; Hospital SG Moscati-Avellino; Istituto Nazionale Tumori-IRCCS "Fondazione G. Pascale" Naples; Hospital AORN Cardarelli Naples; Hospital G Rummo Benevento; Hospital Sant'Anna e San Sebastiano Caserta; University Hospital Università degli Studi di Napoli Federico II Naples; Fondazione Maugeri Benevento; University Hospital San Giovanni di Dio e Ruggi d'Aragona Salerno; Hospital Ospedale dei Colli Naples
| | - Pasquale M Berrino
- The authors would like to thank all the members of the BIO-Cam group who provided patient data for this study: University Hospital of Università degli Studi della Campania "Luigi Vanvitelli" Naples; Hospital SG Moscati-Avellino; Istituto Nazionale Tumori-IRCCS "Fondazione G. Pascale" Naples; Hospital AORN Cardarelli Naples; Hospital G Rummo Benevento; Hospital Sant'Anna e San Sebastiano Caserta; University Hospital Università degli Studi di Napoli Federico II Naples; Fondazione Maugeri Benevento; University Hospital San Giovanni di Dio e Ruggi d'Aragona Salerno; Hospital Ospedale dei Colli Naples
| | - Gabriella di Mauro
- The authors would like to thank all the members of the BIO-Cam group who provided patient data for this study: University Hospital of Università degli Studi della Campania "Luigi Vanvitelli" Naples; Hospital SG Moscati-Avellino; Istituto Nazionale Tumori-IRCCS "Fondazione G. Pascale" Naples; Hospital AORN Cardarelli Naples; Hospital G Rummo Benevento; Hospital Sant'Anna e San Sebastiano Caserta; University Hospital Università degli Studi di Napoli Federico II Naples; Fondazione Maugeri Benevento; University Hospital San Giovanni di Dio e Ruggi d'Aragona Salerno; Hospital Ospedale dei Colli Naples
| | - Liberato Berrino
- The authors would like to thank all the members of the BIO-Cam group who provided patient data for this study: University Hospital of Università degli Studi della Campania "Luigi Vanvitelli" Naples; Hospital SG Moscati-Avellino; Istituto Nazionale Tumori-IRCCS "Fondazione G. Pascale" Naples; Hospital AORN Cardarelli Naples; Hospital G Rummo Benevento; Hospital Sant'Anna e San Sebastiano Caserta; University Hospital Università degli Studi di Napoli Federico II Naples; Fondazione Maugeri Benevento; University Hospital San Giovanni di Dio e Ruggi d'Aragona Salerno; Hospital Ospedale dei Colli Naples
| | - Francesco Rossi
- The authors would like to thank all the members of the BIO-Cam group who provided patient data for this study: University Hospital of Università degli Studi della Campania "Luigi Vanvitelli" Naples; Hospital SG Moscati-Avellino; Istituto Nazionale Tumori-IRCCS "Fondazione G. Pascale" Naples; Hospital AORN Cardarelli Naples; Hospital G Rummo Benevento; Hospital Sant'Anna e San Sebastiano Caserta; University Hospital Università degli Studi di Napoli Federico II Naples; Fondazione Maugeri Benevento; University Hospital San Giovanni di Dio e Ruggi d'Aragona Salerno; Hospital Ospedale dei Colli Naples
| | - Concetta Rafaniello
- The authors would like to thank all the members of the BIO-Cam group who provided patient data for this study: University Hospital of Università degli Studi della Campania "Luigi Vanvitelli" Naples; Hospital SG Moscati-Avellino; Istituto Nazionale Tumori-IRCCS "Fondazione G. Pascale" Naples; Hospital AORN Cardarelli Naples; Hospital G Rummo Benevento; Hospital Sant'Anna e San Sebastiano Caserta; University Hospital Università degli Studi di Napoli Federico II Naples; Fondazione Maugeri Benevento; University Hospital San Giovanni di Dio e Ruggi d'Aragona Salerno; Hospital Ospedale dei Colli Naples
| | - Annalisa Capuano
- The authors would like to thank all the members of the BIO-Cam group who provided patient data for this study: University Hospital of Università degli Studi della Campania "Luigi Vanvitelli" Naples; Hospital SG Moscati-Avellino; Istituto Nazionale Tumori-IRCCS "Fondazione G. Pascale" Naples; Hospital AORN Cardarelli Naples; Hospital G Rummo Benevento; Hospital Sant'Anna e San Sebastiano Caserta; University Hospital Università degli Studi di Napoli Federico II Naples; Fondazione Maugeri Benevento; University Hospital San Giovanni di Dio e Ruggi d'Aragona Salerno; Hospital Ospedale dei Colli Naples
| | | |
Collapse
|
37
|
Arora N, Gupta A, Singh PP. Biological agents in gastrointestinal cancers: adverse effects and their management. J Gastrointest Oncol 2017; 8:485-498. [PMID: 28736636 DOI: 10.21037/jgo.2017.01.07] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Biological therapy comprises agents that by virtue of their unique mechanisms of action, are able to specifically incite a response against or target malignant cells. They differ from conventional chemotherapy with regard to mechanisms of action, indications and side effect profile. Biologic agents have revolutionized therapy for a number of malignancies. In the setting of gastrointestinal (GI) malignancies, agents targeting vascular endothelial growth factor (VEGF), human epidermal growth factor receptor 2 (Her2/Neu) and epidermal growth factor receptor (EGFR) have proven to be invaluable additions to chemotherapy. However, these agents bring with them a set of side effects attributable to their unique mechanisms of action. The anti VEGF agents-bevacizumab, aflibercept and ramucirumab, can result in renal and vascular complications such as hypertension, arterial thrombotic events (ATE), proteinuria and GI perforations. The anti EGFR agents classically cause dermatological toxicities, in addition to hypomagnesemia, which can be dose limiting for patients. Trastuzumab, a monoclonal antibody that targets Her2/Neu, is known to cause cardiotoxicity, especially when used with anthracyclines. Use of immunotherapy agents such as nivolumab is associated with the development immune related adverse events (irAEs). The use of these agents is expected to increase over the next few years and it is crucial that patients and practitioners are aware of their adverse effects and current management strategies. This review highlights the adverse events associated with the use of biologic and immunologic therapies in GI cancers, their incidence and current management strategies.
Collapse
Affiliation(s)
- Nivedita Arora
- Department of Internal Medicine, University of Texas Southwestern Medical Centre, Dallas, TX, USA
| | - Arjun Gupta
- Department of Internal Medicine, University of Texas Southwestern Medical Centre, Dallas, TX, USA
| | - Preet Paul Singh
- Division of Hematology/Oncology, Springfield Clinic Cancer Center, Springfield, Illinois, USA
| |
Collapse
|
38
|
Oral mucosal changes induced by anticancer targeted therapies and immune checkpoint inhibitors. Support Care Cancer 2017; 25:1713-1739. [PMID: 28224235 DOI: 10.1007/s00520-017-3629-4] [Citation(s) in RCA: 104] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Accepted: 02/06/2017] [Indexed: 12/26/2022]
Abstract
Development of biological targeted therapies and immune checkpoint inhibitors has redefined the treatment for many cancers; however, the increasing use of new protocols has led to physicians observing a new spectrum of toxicities. To date, oral adverse events induced by these new anticancer therapies have been mainly reported using nonspecific terminology ("stomatitis," "mucosal inflammation," "mucositis") and remain poorly characterized, with the exception of mammalian target of rapamycin (mTOR) inhibitor-associated stomatitis. Oral toxicities of targeted therapies often display very characteristic features which clearly differ from classic oral injuries observed with cytotoxic chemotherapy and/or radiotherapy. In addition, they frequently affect more than 20% of treated patients and can lead to a significant morbidity or permanent treatment discontinuation. Oral mucosal toxicities described in this review include mTOR inhibitor-associated stomatitis (mIAS); stomatitis, benign migratory glossitis, and osteonecrosis of the jaw associated with multi-targeted kinase inhibitors of the VEGF and PDGF receptors; mucositis induced by EGFR inhibitors (in monotherapy or in combination with head and neck radiotherapy and/or chemotherapy); hyperkeratotic lesions with BRAF inhibitors; pigmentary changes and lichenoid reactions secondary to imatinib; and more recent data on the "Osler-Weber-Rendu-like syndrome" described with the antibody-drug conjugate, TDM-1. Finally, we provide, to our knowledge, the first available structured data on oral toxicities induced by the new recently FDA- and EMA-approved monoclonal antibodies targeting PD-1. Clinical management of these targeted therapy-related oral changes is also discussed.
Collapse
|
39
|
Enhanced tumor-targeting selectivity by modulating bispecific antibody binding affinity and format valence. Sci Rep 2017; 7:40098. [PMID: 28067257 PMCID: PMC5220356 DOI: 10.1038/srep40098] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Accepted: 11/30/2016] [Indexed: 01/07/2023] Open
Abstract
Bispecific antibodies are considered attractive bio-therapeutic agents owing to their ability to target two distinct disease mediators. Cross-arm avidity targeting of antigen double-positive cancer cells over single-positive normal tissue is believed to enhance the therapeutic efficacy, restrict major escape mechanisms and increase tumor-targeting selectivity, leading to reduced systemic toxicity and improved therapeutic index. However, the interplay of factors regulating target selectivity is not well understood and often overlooked when developing clinically relevant bispecific therapeutics. We show in vivo that dual targeting alone is not sufficient to endow selective tumor-targeting, and report the pivotal roles played by the affinity of the individual arms, overall avidity and format valence. Specifically, a series of monovalent and bivalent bispecific IgGs composed of the anti-HER2 trastuzumab moiety paired with affinity-modulated VH and VL regions of the anti-EGFR GA201 mAb were tested for selective targeting and eradication of double-positive human NCI-H358 non-small cell lung cancer target tumors over single-positive, non-target NCI-H358-HER2 CRISPR knock out tumors in nude mice bearing dual-flank tumor xenografts. Affinity-reduced monovalent bispecific variants, but not their bivalent bispecific counterparts, mediated a greater degree of tumor targeting selectivity, while the overall efficacy against the targeted tumor was not substantially affected.
Collapse
|
40
|
Singh A, Venkannagari S, Oh KH, Zhang YQ, Rohde JM, Liu L, Nimmagadda S, Sudini K, Brimacombe KR, Gajghate S, Ma J, Wang A, Xu X, Shahane SA, Xia M, Woo J, Mensah GA, Wang Z, Ferrer M, Gabrielson E, Li Z, Rastinejad F, Shen M, Boxer MB, Biswal S. Small Molecule Inhibitor of NRF2 Selectively Intervenes Therapeutic Resistance in KEAP1-Deficient NSCLC Tumors. ACS Chem Biol 2016; 11:3214-3225. [PMID: 27552339 PMCID: PMC5367156 DOI: 10.1021/acschembio.6b00651] [Citation(s) in RCA: 414] [Impact Index Per Article: 46.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Loss of function mutations in Kelch-like ECH Associated Protein 1 (KEAP1), or gain-of-function mutations in nuclear factor erythroid 2-related factor 2 (NRF2), are common in non-small cell lung cancer (NSCLC) and associated with therapeutic resistance. To discover novel NRF2 inhibitors for targeted therapy, we conducted a quantitative high-throughput screen using a diverse set of ∼400 000 small molecules (Molecular Libraries Small Molecule Repository Library, MLSMR) at the National Center for Advancing Translational Sciences. We identified ML385 as a probe molecule that binds to NRF2 and inhibits its downstream target gene expression. Specifically, ML385 binds to Neh1, the Cap 'N' Collar Basic Leucine Zipper (CNC-bZIP) domain of NRF2, and interferes with the binding of the V-Maf Avian Musculoaponeurotic Fibrosarcoma Oncogene Homologue G (MAFG)-NRF2 protein complex to regulatory DNA binding sequences. In clonogenic assays, when used in combination with platinum-based drugs, doxorubicin or taxol, ML385 substantially enhances cytotoxicity in NSCLC cells, as compared to single agents. ML385 shows specificity and selectivity for NSCLC cells with KEAP1 mutation, leading to gain of NRF2 function. In preclinical models of NSCLC with gain of NRF2 function, ML385 in combination with carboplatin showed significant antitumor activity. We demonstrate the discovery and validation of ML385 as a novel and specific NRF2 inhibitor and conclude that targeting NRF2 may represent a promising strategy for the treatment of advanced NSCLC.
Collapse
Affiliation(s)
- Anju Singh
- Department of Environmental Health Sciences, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland
| | - Sreedhar Venkannagari
- Department of Environmental Health Sciences, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland
| | - Kyu H. Oh
- Department of Environmental Health Sciences, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland
| | - Ya-Qin Zhang
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland
| | - Jason M. Rohde
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland
| | - Li Liu
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland
| | - Sridhar Nimmagadda
- Departments of Radiology and Oncology, School of Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Kuladeep Sudini
- Department of Environmental Health Sciences, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland
| | - Kyle R. Brimacombe
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland
| | - Sachin Gajghate
- Department of Environmental Health Sciences, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland
| | - Jinfang Ma
- Department of Environmental Health Sciences, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland
| | - Amy Wang
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland
| | - Xin Xu
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland
| | - Sampada A. Shahane
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland
| | - Menghang Xia
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland
| | - Juhyung Woo
- Department of Oncology, School of Medicine, Johns Hopkins University, Baltimore, Maryland
| | - George A. Mensah
- Department of Pharmaceutical Sciences, Massachusetts College of Pharmacy and Health Sciences, Worcester, Massachusetts
| | - Zhibin Wang
- Department of Environmental Health Sciences, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland
| | - Marc Ferrer
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland
| | - Edward Gabrielson
- Department of Oncology, School of Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Zhuyin Li
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland
| | - Fraydoon Rastinejad
- Metabolic Signaling and Disease Program, Sanford-Burnham Medical Research Institute, Orlando, Florida
| | - Min Shen
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland
| | - Matthew B. Boxer
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland
| | - Shyam Biswal
- Department of Environmental Health Sciences, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland
- Department of Oncology, School of Medicine, Johns Hopkins University, Baltimore, Maryland
| |
Collapse
|
41
|
Streb J, Püsküllüoğlu M, Glanowska I, Ochenduszko S, Konopka K, Łupkowski R, Michalowska-Kaczmarczyk A, Bochenek-Cibor J, Majka M, Krzemieniecki K. Assessment of frequency and severity of hypomagnesemia in patients with metastatic colorectal cancer treated with cetuximab, with a review of the literature. Oncol Lett 2015; 10:3749-3755. [PMID: 26788202 DOI: 10.3892/ol.2015.3800] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Accepted: 06/16/2015] [Indexed: 01/24/2023] Open
Abstract
Currently, there are a few systemic treatment options for patients with metastatic colorectal cancer (mCRC). Targeted therapy used in this setting includes the use of monoclonal antibodies, such as cetuximab or panitumumab, directed against epidermal growth factor receptor. The aim of the present study was to estimate the frequency and severity of hypomagnesemia among patients with mCRC treated with cetuximab. The data from the Department of Clinical Oncology, University Hospital of Krakow (Krakow, Poland), concerning 52 patients treated between 2009 and 2013 were collected. Of these, 27 patients fulfilled the inclusion criteria to enter this retrospective study. The National Cancer Institute Common Terminology Criteria for Adverse Events version 4.0 were used to grade the level of hypomagnesemia. In total, 29.6% of all patients experienced hypomagnesemia during treatment, and the majority of cases were grade 1 (22.2%). There was no statistically significant correlation between magnesium (Mg) level and patient age, duration of treatment, localization of primary tumor or metastases, and the number of metastases. However, there was an upward trend in a logistic regression model showing that the risk of developing hypomagnesemia increases with age. Hypomagnesemia is a frequent problem among mCRC patients receiving cetuximab. It is essential to introduce guidelines regarding the monitoring of the Mg level and its supplementation in this group of patients.
Collapse
Affiliation(s)
- Joanna Streb
- Department of Clinical Oncology, University Hospital, Jagiellonian University Medical College, Kraków 31-008, Poland
| | - Miroslawa Püsküllüoğlu
- Department of Clinical Oncology, University Hospital, Jagiellonian University Medical College, Kraków 31-008, Poland
| | - Izabela Glanowska
- Department of Clinical Oncology, University Hospital, Jagiellonian University Medical College, Kraków 31-008, Poland
| | - Sebastian Ochenduszko
- Department of Clinical Oncology, University Hospital, Jagiellonian University Medical College, Kraków 31-008, Poland
| | - Kamil Konopka
- Department of Clinical Oncology, University Hospital, Jagiellonian University Medical College, Kraków 31-008, Poland
| | - Radoslaw Łupkowski
- Department of Clinical Oncology, University Hospital, Jagiellonian University Medical College, Kraków 31-008, Poland
| | - Anna Michalowska-Kaczmarczyk
- Department of Clinical Oncology, University Hospital, Jagiellonian University Medical College, Kraków 31-008, Poland
| | | | - Marcin Majka
- Department of Transplantation, Polish-American Institute of Pediatrics, Jagiellonian University Medical College, Kraków 31-008, Poland
| | - Krzysztof Krzemieniecki
- Department of Clinical Oncology, University Hospital, Jagiellonian University Medical College, Kraków 31-008, Poland
| |
Collapse
|
42
|
Holcmann M, Sibilia M. Mechanisms underlying skin disorders induced by EGFR inhibitors. Mol Cell Oncol 2015; 2:e1004969. [PMID: 27308503 PMCID: PMC4905346 DOI: 10.1080/23723556.2015.1004969] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Revised: 12/31/2014] [Accepted: 01/03/2015] [Indexed: 12/21/2022]
Abstract
The epidermal growth factor receptor (EGFR) is a receptor tyrosine kinase that is frequently mutated or overexpressed in a large number of tumors such as carcinomas or glioblastoma. Inhibitors of EGFR activation have been successfully established for the therapy of some cancers and are more and more frequently being used as first or later line therapies. Although the side effects induced by inhibitors of EGFR are less severe than those observed with classic cytotoxic chemotherapy and can usually be handled by out-patient care, they may still be a cause for dose reduction or discontinuation of treatment that can reduce the effectiveness of antitumor therapy. The mechanisms underlying these cutaneous side effects are only partly understood. Important questions, such as the reasons for the correlation between the intensity of the side effects and the efficiency of treatment with EGFR inhibitors, remain to be answered. Optimized adjuvant strategies to accompany anti-EGFR therapy need to be found for optimal therapeutic application and improved quality of life of patients. Here, we summarize current literature on the molecular and cellular mechanisms underlying the cutaneous side effects induced by EGFR inhibitors and provide evidence that keratinocytes are probably the optimal targets for adjuvant therapy aimed at alleviating skin toxicities.
Collapse
Affiliation(s)
- Martin Holcmann
- Institute of Cancer Research; Department of Medicine I; Medical University of Vienna; Comprehensive Cancer Center ; Vienna, Austria
| | - Maria Sibilia
- Institute of Cancer Research; Department of Medicine I; Medical University of Vienna; Comprehensive Cancer Center ; Vienna, Austria
| |
Collapse
|
43
|
Epidermal growth factor receptor inhibitors: a review of cutaneous adverse events and management. Dermatol Res Pract 2014; 2014:734249. [PMID: 24723942 PMCID: PMC3958662 DOI: 10.1155/2014/734249] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2013] [Accepted: 01/23/2014] [Indexed: 12/14/2022] Open
Abstract
Epidermal growth factor inhibitors (EGFRI), the first targeted cancer therapy, are currently an essential treatment for many advance-stage epithelial cancers. These agents have the superior ability to target cancers cells and better safety profile compared to conventional chemotherapies. However, cutaneous adverse events are common due to the interference of epidermal growth factor receptor (EGFR) signaling in the skin. Cutaneous toxicities lead to poor compliance, drug cessation, and psychosocial discomfort. This paper summarizes the current knowledge concerning the presentation and management of skin toxicity from EGFRI. The common dermatologic adverse events are papulopustules and xerosis. Less common findings are paronychia, regulatory abnormalities of hair growth, maculopapular rash, mucositis, and postinflammatory hyperpigmentation. Radiation enhances EGFRI rash due to synergistic toxicity. There is a positive correlation between the occurrence and severity of cutaneous adverse effects and tumor response. To date, prophylactic systemic tetracycline and tetracycline class antibiotics have proven to be the most effective treatment regime.
Collapse
|
44
|
Passaro A, Cortesi E, de Marinis F. Second-line treatment of non-small-cell lung cancer: chemotherapy or tyrosine kinase inhibitors? Expert Rev Anticancer Ther 2014; 11:1587-97. [DOI: 10.1586/era.11.120] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
45
|
Hocking CM, Price TJ. Panitumumab in the management of patients with KRAS wild-type metastatic colorectal cancer. Therap Adv Gastroenterol 2014; 7:20-37. [PMID: 24381645 PMCID: PMC3871277 DOI: 10.1177/1756283x13498660] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
The past 15 years has seen a marked increase in available therapeutic options for patients with metastatic colorectal cancer resulting in improvements in median survival from 12 to 24 months. One of these new options is panitumumab, which is a fully humanized monoclonal antibody that binds to the epidermal growth factor receptor of tumor cells and inhibits downstream cell signaling with antitumor effects of inhibition of tumor growth, induction of apoptosis and inhibition of angiogenesis. Large randomized clinical trials have demonstrated significant improvements in tumor response rates and progression-free survival when panitumumab is combined with chemotherapy and as monotherapy in chemorefractory metastatic colorectal cancer. Clinical benefit with panitumumab is limited to patients with nonmutated KRAS tumors. Rash is a common toxicity of panitumumab treatment but can potentially be ameliorated with the use of prophylactic strategies. The role of panitumumab in the overall treatment of metastatic colorectal cancer is evolving and future clinical trials will focus on improved patient selection through use of novel predictive biomarkers, and the optimal timing of treatment.
Collapse
Affiliation(s)
- Christopher M Hocking
- Department of Medical Oncology, The Queen Elizabeth Hospital, Woodville, SA, Australia
| | - Timothy J Price
- Department of Medical Oncology, TQEH, Woodville, Woodville Road, Woodville, SA 5011, Australia
| |
Collapse
|
46
|
Leporini C, Saullo F, Filippelli G, Sorrentino A, Lucia M, Perri G, Gattuta GL, Infusino S, Toscano R, Dima G, Olivito V, Paletta L, Bottoni U, De Sarro G. Management of dermatologic toxicities associated with monoclonal antibody epidermal growth factor receptor inhibitors: A case review. J Pharmacol Pharmacother 2013; 4:S78-85. [PMID: 24347989 PMCID: PMC3853676 DOI: 10.4103/0976-500x.120966] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
INTRODUCTION The epidermal growth factor receptor inhibitors (EGFRIs), cetuximab and panitumumab, represent an effective treatment option for patients affected by metastatic colorectal cancer (mCRC); furthermore, they are relatively devoid of systemic toxicities, which are commonly observed with standard cytotoxic chemotherapy. However, the majority of patients treated with these monoclonal antibodies (mAbs), will experience dermatologic toxicities, most notably the papulopustular skin rash, which can impact quality-of-life and affect adherence to therapy. This paper reviews the most recent practices in the management of skin rash related to anti-epidermal growth factor receptor (EGFR) mAbs, cetuximab and panitumumab, in the treatment of mCRC. MATERIALS AND METHODS We reviewed relevant literature regarding dermatologic toxicities associated with anti-EGFR mAbs in order to give important indications about prevention and reactive treatment of skin rash. RESULTS Two case reports were presented to show how skin rash could hamper mAb EGFRIs use in clinical practice, underscoring the need of implementing a comprehensive management strategy of skin toxicity in order to promote patients' compliance with anti-EGFR therapy and maintain quality-of-life. Based on randomized data, recent guidelines established by the Multinational Association for Supportive Care in Cancer Skin Toxicity Study Group suggest that prophylactic use of oral doxycycline or minocycline reduces the risk and severity of skin rash, improving clinical outcomes. CONCLUSIONS At the start of treatment with cetuximab and panitumumab, the proper patient education about the skin rash associated with these mAbs and the implementation of a pre-emptive, comprehensive skin toxicity program significantly contribute to improve adherence to therapy, optimize anti-EGFR therapy and maintain quality-of-life.
Collapse
Affiliation(s)
- Christian Leporini
- Department of Science of Health, School of Medicine, University of Catanzaro, Catanzaro, Italy
- Pharmacovigilance's Centre Calabria Region, University Hospital Mater Domini, Catanzaro, Italy
| | - Francesca Saullo
- Department of Science of Health, School of Medicine, University of Catanzaro, Catanzaro, Italy
- Pharmacovigilance's Centre Calabria Region, University Hospital Mater Domini, Catanzaro, Italy
| | | | - Antonio Sorrentino
- Pharmacy Unit, S. Francesco di Paola Hospital, Paola Province of Cosenza, Italy
| | - Maria Lucia
- Oncology Unit, S. Francesco di Paola Hospital, Paola Province of Cosenza, Italy
| | - Gino Perri
- Oncology Unit, S. Francesco di Paola Hospital, Paola Province of Cosenza, Italy
| | - Gaetana La Gattuta
- Oncology Unit, S. Francesco di Paola Hospital, Paola Province of Cosenza, Italy
| | - Stefania Infusino
- Oncology Unit, S. Francesco di Paola Hospital, Paola Province of Cosenza, Italy
| | - Rosa Toscano
- Oncology Unit, S. Francesco di Paola Hospital, Paola Province of Cosenza, Italy
| | - Gianluca Dima
- Oncology Unit, S. Francesco di Paola Hospital, Paola Province of Cosenza, Italy
| | - Virginia Olivito
- Oncology Unit, S. Francesco di Paola Hospital, Paola Province of Cosenza, Italy
| | - Laura Paletta
- Department of Science of Health, School of Medicine, University of Catanzaro, Catanzaro, Italy
- Pharmacovigilance's Centre Calabria Region, University Hospital Mater Domini, Catanzaro, Italy
| | - Ugo Bottoni
- Department of Science of Health, School of Medicine, University of Catanzaro, Catanzaro, Italy
- Pharmacovigilance's Centre Calabria Region, University Hospital Mater Domini, Catanzaro, Italy
| | - Giovambattista De Sarro
- Department of Science of Health, School of Medicine, University of Catanzaro, Catanzaro, Italy
- Pharmacovigilance's Centre Calabria Region, University Hospital Mater Domini, Catanzaro, Italy
| |
Collapse
|
47
|
Azizi E, Kittai A, Kozuch P. Antiepidermal growth factor receptor monoclonal antibodies: applications in colorectal cancer. CHEMOTHERAPY RESEARCH AND PRACTICE 2012; 2012:198197. [PMID: 23091721 PMCID: PMC3472558 DOI: 10.1155/2012/198197] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/16/2012] [Accepted: 06/12/2012] [Indexed: 01/19/2023]
Abstract
Patients with metastatic colorectal cancer have a poor prognosis and present a challenge to clinicians. The role of the antiepidermal growth factor receptor (EGFR) pathway in tumorogenesis and tumor progression has been well defined. This paper will review the use of anti-EGFR monoclonal antibodies in the treatment of operable, as well as metastatic colorectal cancer both in the setting of KRAS mutation unselected patients and later in KRAS wild-type patients. Active investigations designed to further identify predictive biomarkers that may be potentially druggable are reviewed as well.
Collapse
Affiliation(s)
- Efat Azizi
- Beth Israel Medical Center, Phillips Ambulatory Care Center, Continuum Cancer Centers of New York, 10 Union Square East, Suite 4C, New York, NY, USA ; Section of Hematology/Oncology, Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
| | | | | |
Collapse
|
48
|
Leon L, Vázquez S, Gracia JM, Casal J, Lazaro M, Firvida JL, Amenedo M, Santome L, Macia S. First-line bevacizumab, cisplatin and vinorelbine plus maintenance bevacizumab in advanced non-squamous non-small cell lung cancer chemo-naïve patients. Expert Opin Pharmacother 2012; 13:1389-96. [PMID: 22630129 DOI: 10.1517/14656566.2012.693165] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
OBJECTIVE The aim of this study was to evaluate efficacy and safety of first-line treatment with bevacizumab, cisplatin and vinorelbine and bevacizumab maintenance in non-squamous, non-small cell lung cancer (NSCLC). RESEARCH DESIGN AND METHODS Forty-nine patients with stage IIIB plus pleural effusion or stage IV NSCLC were included in a Phase II clinical trial. Treatment consisted of 3-week cycles of bevacizumab (15 mg/kg on day 1), cisplatin (80 mg/m(2) on day 1) and vinorelbine (25 mg/m(2) on days 1 and 8). After 6 cycles, non-progressing patients received bevacizumab maintenance therapy. The primary end point was progression-free survival (PFS), calculated using the Kaplan-Meier method. RESULTS Thirteen (29%) of 45 evaluable patients presented a partial response. PFS and overall survival were 6.0 months (95% confidence interval (CI) 4.5 - 7.5) and 14.7 months (95% CI 8.4 - 21), respectively. Fourteen patients (28%) experienced grade 3 - 4 neutropenia and 7 (14%) experienced febrile neutropenia during the combination treatment. During the maintenance phase, the most frequent grade 3 - 4 adverse event was hypertension. Neither grade 3 - 4 thrombocytopenia nor toxic death was observed. CONCLUSIONS The studied regimen achieved a similar efficacy to other regimens containing platinum doublets. The data provide further evidence that bevacizumab may be used in combination with multiple standard platinum-based doublets in this setting.
Collapse
Affiliation(s)
- Luis Leon
- Complexo Hospitalario Universitario de Santiago, Medical Oncology Department, Travesía da Choupana, s/n, 15706 Santiago de Compostela, Spain.
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Neeff HP, Drognitz O, Klock A, Illerhaus G, Opitz OG, Hopt UT, Makowiec F. Impact of preoperative targeted therapy on postoperative complications after resection of colorectal liver metastases. Int J Colorectal Dis 2012; 27:635-45. [PMID: 22139030 DOI: 10.1007/s00384-011-1360-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/10/2011] [Indexed: 02/06/2023]
Abstract
PURPOSE The impact of chemotherapy (CTx) on morbidity after liver resection for colorectal metastases (CRC-LM) has been increasingly investigated during recent years. Biologic agents like bevacizumab (BEV) or cetuximab (CET) are now added as "targeted therapy" (TT), also in neoadjuvant settings. Initial series could demonstrate the safety of those regimens in liver resection but data are still scarce. We evaluated the impact of CTx with BEV or CET (CTx + TT) on perioperative morbidity and mortality. METHODS Two hundred thirty-seven patients who underwent liver resections for CRC-LM after chemotherapy before surgery since 1999 were included. One hundred eighty-five patients (78%) had preoperative CTx regimen without biologic agents (fluoropyrimidine-, oxaliplatin-, or irinotecan-based) and 52 (22%) had CTx + TT (39 BEV, 11 CET, 2 CET/BEV). After preoperative CTx + TT, a time interval of at least 4-6 weeks and a residual liver volume of >35% before surgery were required. RESULTS Hemihepatectomy or more was performed in about half of the patients. The median amount of intraoperatively transfused blood was 0 ml in both groups (p = 0.34). Overall mortality was 1.7% and slightly elevated in patients with CTx + TT (3.8% vs. 1.1%, p = 0.17). Any complication occurred in (CTx + TT vs. CTx) 52% and 46%, respectively (p = 0.47). The rates of liver failure (9.6% vs. 9.7%, p = 0.98), infectious complications such as wound infection (19% vs. 16%, p = 0.62) and abdominal abscess (8% vs. 6.5%, p = 0.71), as well as the rate of relaparotomies (11.5% vs. 7.0%, p = 0.29) showed no significant differences between the groups with TT or without. In multivariate analyses, neither type nor duration of CTx nor the time interval between CTx and surgery showed any influence on complication rates. CONCLUSIONS Our data confirm the safety of targeted therapy before liver resection for CRC-LM. This effect may in part be due to our treatment policy (time interval to resection and residual liver volume) after intensive preoperative CTx.
Collapse
Affiliation(s)
- Hannes P Neeff
- Department of Surgery, University of Freiburg, Hugstetter Strasse 55, 79106 Freiburg, Germany.
| | | | | | | | | | | | | |
Collapse
|
50
|
Wang J, Li K, Wang B, Bi J. Lymphatic microvessel density as a prognostic factor in non-small cell lung carcinoma: a meta-analysis of the literature. Mol Biol Rep 2011; 39:5331-8. [PMID: 22167333 DOI: 10.1007/s11033-011-1332-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2011] [Accepted: 12/03/2011] [Indexed: 01/27/2023]
Abstract
The role of lymphatic microvessel density (LVD) as a prognostic factor for survival of patients with non-small cell lung carcinoma (NSCLC) remains controversial. To evaluate this potential role, we performed a systematic review of the electronic databases PubMed and EMBASE for relevant literature to review and compile available survival results. To be eligible, a study had to assess LVD in patients with NSCLC and to compare survival based on LVD stratification. Among 12 eligible trials, all dealt with NSCLC, and 10 trials provided results for the meta-analysis of survival data (evaluable trials). In terms of survival, high LVD was reported to be an unfavorable prognostic factor for overall survival in 8 studies, whereas it was not in 4 studies. The overall survival hazard ratio for the 10 evaluable studies (1,426 patients) was calculated to be 1.41 (95% CI: 1.14-1.75) using a random effects model, indicating a poorer survival for NSCLC patients with high LVD. The hazard ratio was 1.52 (95% CI: 1.10-2.11) in 5 NSCLC studies where LVD was assessed based on D2-40 and 1.31 (95% CI: 1.08-1.60) in 4 studies where LVD was measured based on vascular endothelial growth factor receptor-3. This study supports the hypothesis that the lymphatic microvessel count or LVD, which reflects levels of lymphangiogenesis, is a poor prognostic factor for patient survival in surgically treated NSCLC. However, the present findings may overestimate the prognostic capacity of LVD because of publication and report bias. In addition, the standardization of lymphangiogenesis assessment by the lymphatic microvessel count is necessary.
Collapse
Affiliation(s)
- Jun Wang
- Department of Oncology, General Hospital, Jinan Command of People's Liberation Army, Shifan Street 25, Tianqiao District, Jinan, 250031, China
| | | | | | | |
Collapse
|