1
|
Alkhateeb MA, Aljarba NH, Yousafi Q, Anwar F, Biswas P. Elucidating gastric cancer mechanisms and therapeutic potential of Adociaquinone A targeting EGFR: A genomic analysis and Computer Aided Drug Design (CADD) approach. J Cell Mol Med 2024; 28:e70133. [PMID: 39434198 PMCID: PMC11493557 DOI: 10.1111/jcmm.70133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 05/05/2024] [Accepted: 09/09/2024] [Indexed: 10/23/2024] Open
Abstract
Gastric cancer predominantly adenocarcinoma, accounts for over 85% of gastric cancer diagnoses. Current therapeutic options are limited, necessitating the discovery of novel drug targets and effective treatments. The Affymetrix gene expression microarray dataset (GSE64951) was retrieved from NCBI-GEO data normalization and DEGs identification was done by using R-Bioconductor package. Gene Ontology (GO) analysis of DEGs was performed using DAVID. The protein-protein interaction network was constructed by STRING database plugin in Cytoscape. Subclusters/modules of important interacting genes in main network were extracted by using MCODE. The hub genes from in the network were identified by using Cytohubba. The miRNet tool built a hub gene/mRNA-miRNA network and Kaplan-Meier-Plotter conducted survival analysis. AutoDock Vina and GROMACS MD simulations were used for docking and stability analysis of marine compounds against the 5CNN protein. Total 734 DEGs (507 up-regulated and 228 down-regulated) were identified. Differentially expressed genes (DEGs) were enriched in processes like cell-cell adhesion and ATP binding. Eight hub genes (EGFR, HSPA90AA1, MAPK1, HSPA4, PPP2CA, CDKN2A, CDC20, and ATM) were selected for further analysis. A total of 23 miRNAs associated with hub genes were identified, with 12 of them targeting PPP2CA. EGFR displayed the highest expression and hazard rate in survival analyses. The kinase domain of EGFR (PDBID: 5CNN) was chosen as the drug target. Adociaquinone A from Petrosia alfiani, docked with 5CNN, showed the lowest binding energy with stable interactions across a 50 ns MD simulation, highlighting its potential as a lead molecule against EGFR. This study has identified crucial DEGs and hub genes in gastric cancer, proposing novel therapeutic targets. Specifically, Adociaquinone A demonstrates promising potential as a bioactive drug against EGFR in gastric cancer, warranting further investigation. The predicted miRNA against the hub gene/proteins can also be used as potential therapeutic targets.
Collapse
Affiliation(s)
| | - Nada H. Aljarba
- Department of Biology, College of SciencePrincess Nourah bint Abdulrahman UniversityRiyadhSaudi Arabia
| | - Qudsia Yousafi
- Department of BiosciencesCOMSATS University Islamabad, Sahiwal CampusSahiwalPakistan
| | - Fatima Anwar
- Department of BiosciencesCOMSATS University Islamabad, Sahiwal CampusSahiwalPakistan
| | - Partha Biswas
- Laboratory of Pharmaceutical Biotechnology and Bioinformatics, Department of Genetic Engineering and BiotechnologyJashore University of Science and TechnologyJashoreBangladesh
| |
Collapse
|
2
|
Mehrotra S, Kupani M, Kaur J, Kaur J, Pandey RK. Immunotherapy guided precision medicine in solid tumors. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2024; 140:249-292. [PMID: 38762271 DOI: 10.1016/bs.apcsb.2024.02.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2024]
Abstract
Cancer is no longer recognized as a single disease but a collection of diseases each with its defining characteristics and behavior. Even within the same cancer type, there can be substantial heterogeneity at the molecular level. Cancer cells often accumulate various genetic mutations and epigenetic alterations over time, leading to a coexistence of distinct subpopulations of cells within the tumor. This tumor heterogeneity arises not only due to clonal outgrowth of cells with genetic mutations, but also due to interactions of tumor cells with the tumor microenvironment (TME). The latter is a dynamic ecosystem that includes cancer cells, immune cells, fibroblasts, endothelial cells, stromal cells, blood vessels, and extracellular matrix components, tumor-associated macrophages and secreted molecules. The complex interplay between tumor heterogeneity and the TME makes it difficult to develop one-size-fits-all treatments and is often the cause of therapeutic failure and resistance in solid cancers. Technological advances in the post-genomic era have given us cues regarding spatial and temporal tumor heterogeneity. Armed with this knowledge, oncologists are trying to target the unique genomic, epigenetic, and molecular landscape in the tumor cell that causes its oncogenic transformation in a particular patient. This has ushered in the era of personalized precision medicine (PPM). Immunotherapy, on the other hand, involves leveraging the body's immune system to recognize and attack cancer cells and spare healthy cells from the damage induced by radiation and chemotherapy. Combining PPM and immunotherapy represents a paradigm shift in cancer treatment and has emerged as a promising treatment modality for several solid cancers. In this chapter, we summarise major types of cancer immunotherapy and discuss how they are being used for precision medicine in different solid tumors.
Collapse
Affiliation(s)
- Sanjana Mehrotra
- Department of Human Genetics, Guru Nanak Dev University, Amritsar, Punjab, India.
| | - Manu Kupani
- Department of Human Genetics, Guru Nanak Dev University, Amritsar, Punjab, India
| | - Jaismeen Kaur
- Department of Human Genetics, Guru Nanak Dev University, Amritsar, Punjab, India
| | - Jashandeep Kaur
- Department of Human Genetics, Guru Nanak Dev University, Amritsar, Punjab, India
| | - Rajeev Kumar Pandey
- Research and Development-Protein Biology, Thermo Fisher Scientific, Bengaluru, Karnataka, India
| |
Collapse
|
3
|
Bokemeyer C, Ciardiello F, Dubreuil O, Guigay J, Kasper S, Pfeiffer P, Pinto C, Yamaguchi K, Yoshino T, Zielinski C, Esser R, Tabernero J. Cetuximab every 2 weeks versus standard weekly dosing administration schedule. Future Oncol 2024; 20:393-407. [PMID: 37850363 DOI: 10.2217/fon-2023-0282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2023] Open
Abstract
Cetuximab every 2 weeks (Q2W) dosing schedule is approved by the US FDA and by the Japanese Pharmaceuticals and Medical Devices Agency in patients with metastatic colorectal cancer and squamous cell carcinoma of the head and neck. Phase II trials have found comparable efficacy and safety for the weekly (Q1W) and Q2W schedules, and real-world studies have shown noninferiority of the Q2W compared with the Q1W schedule. Several guidelines recommend cetuximab Q2W administration as an alternative to the Q1W dosing schedule. Cetuximab Q2W can be administered with a Q2W dose of chemotherapy, making it a more convenient option to the Q1W schedule, potentially resulting in reduced costs for administration, increased flexibility for clinical staff and improved patient adherence.
Collapse
Affiliation(s)
- Carsten Bokemeyer
- The II Medical Clinic, Department of Oncology, Hematology & BMT with section of Pneumology, University Medical Center of Hamburg-Eppendorf, Hamburg, Germany
| | - Fortunato Ciardiello
- Department of Precision Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Olivier Dubreuil
- Medical Oncology Unit, Diaconesses-Croix St Simon Hospital, Paris, France
| | - Joel Guigay
- Groupe d'Oncologie Radiothérapie Tête Et Cou (GORTEC), Tours, France
| | - Stefan Kasper
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen, Essen, Germany
| | - Per Pfeiffer
- Department of Oncology, Odense University Hospital, Odense, Denmark
| | - Carmine Pinto
- Medical Oncology, Comprehensive Cancer Center, AUSL-IRCCS of Reggio Emilia, Reggio Emilia, Italy
| | - Kensei Yamaguchi
- Cancer Institute Hospital of the Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Takayuki Yoshino
- Department of Gastroenterology & Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | | | | | - Josep Tabernero
- Medical Oncology Department, Vall d'Hebron Hospital Campus & Institute of Oncology (VHIO), IOB-Quiron, UVic-UCC, Barcelona, Spain
| |
Collapse
|
4
|
Yip PL, Fung WHB, Lee FAS, Lee CF, Wong NSM, Lee SF. Effectiveness and safety of capecitabine, irinotecan and panitumumab in advanced colorectal cancer. Front Oncol 2023; 13:1138357. [PMID: 37091154 PMCID: PMC10116611 DOI: 10.3389/fonc.2023.1138357] [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: 01/10/2023] [Accepted: 03/06/2023] [Indexed: 04/08/2023] Open
Abstract
Introduction Capecitabine, irinotecan, and panitumumab (CAPIRI-P) is a controversial regimen for metastatic colorectal cancer, with concerns regarding the efficacy and toxicity. However, its toxicity profile has been improved with dose reduction, and concerns regarding efficacy have been extrapolated from other trials. This retrospective study reports the real-world effectiveness and safety of modified CAPIRI-P (mCAPIRI-P). Material and methods Advanced colorectal cancer patients receiving mCAPIPI-P in the first-line setting between July 2019 and December 2021 were analyzed. The progression-free survival on treatment (PFSOT) and overall survival (OS) were estimated using the Kaplan-Meier method, and the association with clinical and disease factors was analyzed using the Cox regression model. Serial changes in carcinoembryonic antigen (CEA) level and treatment toxicity were also evaluated. Results A total of 106 patients were included, of whom 97 (92%) and 31 (29%) had left-sided primary and unresectable liver-only disease, respectively. The median PFSOT and OS were 15.4 (95% CI 12.5-18.3) and 25.5 (95% CI 17.6-33.4) months, respectively. Sixteen (51.6%) and 10 (32.3%) liver-only disease patients underwent secondary liver treatment and R0 resection, respectively. In multivariable Cox regression, CEA responders (PFSOT: HR 0.53) and CEA normalization (PFSOT: HR 0.27; OS: HR 0.28) were independent favorable prognostic factors for PFSOT and OS. Grade ≥3 toxicity rate was 43%, mainly related to uncomplicated hematological toxicities. Conclusion The real-world data show that mCAPIRI-P is safe and effective as the first-line treatment regimen for RAS wild-type advanced colorectal cancer and warrants further study.
Collapse
Affiliation(s)
- Pui Lam Yip
- Department of Clinical Oncology, Tuen Mun Hospital, New Territories West Cluster, Hospital Authority, Hong Kong, Hong Kong SAR, China
- Department of Radiation Oncology, National University Cancer Institute, National University Hospital, Singapore, Singapore
| | - Wai Him Brian Fung
- Department of Radiology and Nuclear Medicine, Tuen Mun Hospital, New Territories West Cluster, Hospital Authority, Hong Kong, Hong Kong SAR, China
| | - Francis Ann Shing Lee
- Department of Clinical Oncology, Tuen Mun Hospital, New Territories West Cluster, Hospital Authority, Hong Kong, Hong Kong SAR, China
| | - Chak Fei Lee
- Department of Pharmacy, Tuen Mun Hospital, New Territories West Cluster, Hospital Authority, Hong Kong, Hong Kong SAR, China
| | - Natalie Sean Man Wong
- Department of Clinical Oncology, Tuen Mun Hospital, New Territories West Cluster, Hospital Authority, Hong Kong, Hong Kong SAR, China
| | - Shing Fung Lee
- Department of Clinical Oncology, Tuen Mun Hospital, New Territories West Cluster, Hospital Authority, Hong Kong, Hong Kong SAR, China
- Department of Radiation Oncology, National University Cancer Institute, National University Hospital, Singapore, Singapore
| |
Collapse
|
5
|
Saoudi Gonzalez N, López D, Gómez D, Ros J, Baraibar I, Salva F, Tabernero J, Élez E. Pharmacokinetics and pharmacodynamics of approved monoclonal antibody therapy for colorectal cancer. Expert Opin Drug Metab Toxicol 2022; 18:755-767. [PMID: 36582117 DOI: 10.1080/17425255.2022.2160316] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
INTRODUCTION The introduction of monoclonal antibodies to the chemotherapy backbone treatment has challenged the paradigm of metastatic colorectal cancer (mCRC) treatment. Their mechanism of action and pharmacokinetics are complex but important to understand in order to improve patient selection and treatment outcomes for mCRC population. AREAS COVERED This review examines the scientific data, pharmacodynamics, and pharmacokinetics of approved monoclonal antibodies used to treat mCRC patients, including agents targeting signaling via VEGFR (bevacizumab and ramucirumab), EGFR (cetuximab and panitumumab), HER2/3 target therapy, and immunotherapy agents such as pembrolizumab or nivolumab. Efficacy and mechanism of action of bispecific antibodies are also covered. EXPERT OPINION mCRC is a heterogeneous disease and the optimal selection and sequence of treatments is challenging. Monoclonal antibodies have complex pharmacokinetics and pharmacodynamics, with important interactions between them. The arrival of bioequivalent molecules to the market increases the need for the characterization of pharmacokinetics and pharmacodynamics of classic monoclonal antibodies to reach bioequivalent novel molecules.
Collapse
Affiliation(s)
- Nadia Saoudi Gonzalez
- Medical Oncology Department, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain.,Medical Oncology Department, Vall d'Hebron Institute of Oncology, Vhio Barcelona, Spain
| | - Daniel López
- Medical Oncology Department, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Diego Gómez
- Medical Oncology Department, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Javier Ros
- Medical Oncology Department, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain.,Medical Oncology Department, Vall d'Hebron Institute of Oncology, Vhio Barcelona, Spain
| | - Iosune Baraibar
- Medical Oncology Department, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain.,Medical Oncology Department, Vall d'Hebron Institute of Oncology, Vhio Barcelona, Spain
| | - Francesc Salva
- Medical Oncology Department, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain.,Medical Oncology Department, Vall d'Hebron Institute of Oncology, Vhio Barcelona, Spain
| | - Josep Tabernero
- Medical Oncology Department, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain.,Medical Oncology Department, Vall d'Hebron Institute of Oncology, Vhio Barcelona, Spain
| | - Elena Élez
- Medical Oncology Department, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain.,Medical Oncology Department, Vall d'Hebron Institute of Oncology, Vhio Barcelona, Spain
| |
Collapse
|
6
|
Zhang W, Han X, Yang L, Song Y, Xie L, Gai W, Wang Y, Shi Y. Safety, pharmacokinetics and efficacy of SCT200, an anti-EGFR monoclonal antibody in patients with wild-type KRAS/NRAS/BRAF metastatic colorectal cancer: a phase I dose-escalation and dose-expansion study. BMC Cancer 2022; 22:1104. [PMID: 36307775 PMCID: PMC9617324 DOI: 10.1186/s12885-022-10147-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 09/26/2022] [Indexed: 12/02/2022] Open
Abstract
Background An over-expression of the epidermal growth factor receptor (EGFR) has been observed in colorectal cancer and is associated with aggressive disease and poor prognosis. SCT200 is a newly developed recombinant, fully humanized, anti-EGFR monoclonal antibody. This study aimed to evaluate its safety, tolerability, pharmacokinetics (PK), and efficacy in patients with wild-type KRAS/NRAS/BRAF metastatic colorectal cancer (mCRC). Methods This phase I study comprising dose-escalation phase and dose-expansion phase. SCT200 was administrated intravenously to groups of three to six patients. An every 3-week dosing cycle (0.5–15.0 mg/kg) and multiple dosing schedule were evaluated. Blood samples were collected at preset intervals for PK assessment, radiological imaging was used for efficacy assessment, and continuous safety monitoring was performed in each group during the study. Results From December 16, 2014 to December 31, 2018, fifty-six patients with wild-type KRAS/NRAS/BRAF mCRC receiving ≥ 1 dose of SCT200 were evaluated. Among them, 44.6% (25/56) of the patients failed at least two prior lines of chemotherapy. No dose-limiting toxicities occurred in any group. All of the patients experienced treatment-emergent adverse events (TEAEs). 96.4% (54/56) of patients experienced treatment-related adverse events (TRAEs), and 26.8% (15/56) of patients with Grade ≥ 3 TRAEs. No serious TRAEs were observed. The most common TRAEs were dermotoxicity and hypomagnesemia. PK analysis showed non-linear PK in the range of 0.5 - 8.0 mg/kg of single dose SCT200, the clearance decreased, and the elimination half-life (T1/2) prolonged following dose increase. In the multiple-dose period, the clearance decreased, peak concentration increased, and T1/2 prolonged during prolonged drug administration, and a steady state was reached after five consecutive dose of 6.0 mg/kg quaque week (QW). The objective response rate (ORR) was 30.4% (17/56, 95% confidence interval [CI], 18.8%–44.1%). The ORR in the dose-expansion group (6.0 mg/kg QW) was 48.0% (12/25, 95% CI, 27.8%–68.7%), the median progression-free survival was 5.2 months (95%CI, 3.6–5.5), and the median overall survival was 20.2 months (95%CI, 12.1-not reached). Conclusions SCT200 showed favorable safety, PK profile, and preliminary efficacy for patients with wild-type KRAS/NRAS/BRAF mCRC. Trial registration This study was registered with ClinicalTrials.gov (NCT02211443). Supplementary Information The online version contains supplementary material available at 10.1186/s12885-022-10147-9.
Collapse
|
7
|
FBXW2 inhibits prostate cancer proliferation and metastasis via promoting EGFR ubiquitylation and degradation. Cell Mol Life Sci 2022; 79:268. [PMID: 35499593 PMCID: PMC9061686 DOI: 10.1007/s00018-022-04320-3] [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: 01/08/2022] [Revised: 04/17/2022] [Accepted: 04/20/2022] [Indexed: 11/03/2022]
Abstract
FBXW2 is a poorly characterized F-box protein, as a tumor suppressor that inhibits growth and metastasis of lung cancer by promoting ubiquitylation and degradation of oncogenic proteins, including SKP2 and β-catenin. However, what the biological functions of FBXW2 in prostate cancer cells and whether FBXW2 targets other substrates to involve in progression of prostate cancer is still unclear. Here, we reported that overexpression of FBXW2 attenuated proliferation and metastasis of PCa models both in vitro and in vivo, while FBXW2 depletion exhibited the opposite effects. Intriguingly, FBXW2 was an E3 ligase for EGFR in prostate cancer. EGFR protein level and its half-life were extended by FBXW2 depletion, while EGFR protein level was decreased, and its half-life was shortened upon overexpression of FBXW2, but not its dominant-negative mutant. Importantly, FBXW2 bond to EGFR via its consensus degron motif (TSNNST), and ubiquitylated and degraded EGFR, resulting in repression of EGF function. Thus, our data uncover a novel that FBXW2 as a tumor suppressor of prostate cancer, inhibits EGFR downstream by promoting EGFR ubiquitination and degradation, resulting in repression of cell proliferation and metastasis.
Collapse
|
8
|
Bai M, Wang M, Deng T, Bai Y, Zang K, Miao Z, Gai W, Xie L, Ba Y. Safety and efficacy of anti-EGFR monoclonal antibody (SCT200) as second-line therapy in advanced esophageal squamous cell carcinoma. Cancer Biol Med 2022; 19:j.issn.2095-3941.2021.0388. [PMID: 35014769 PMCID: PMC8958882 DOI: 10.20892/j.issn.2095-3941.2021.0388] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Accepted: 10/21/2021] [Indexed: 12/24/2022] Open
Abstract
OBJECTIVE The mainstay treatment of esophageal squamous cell carcinoma (ESCC) involves chemotherapy and immunotherapy. However, alternative therapies are required for patients who are refractory or intolerant to existing therapies. METHODS In this single-arm, multicenter, open-label phase Ib study, 30 patients received an intravenous infusion of SCT200, an antiepidermal growth factor receptor (EGFR) monoclonal antibody, 6.0 mg/kg once a week for 6 weeks, followed by 8.0 mg/kg once every 2 weeks until disease progression or intolerable toxicity. The primary endpoint was the objective response rate (ORR). The secondary endpoints were progression-free survival (PFS), overall survival (OS), and safety. RESULTS Thirty patients were enrolled between July 2018 and May 2019. The ORR was 16.7% (95% CI: 5.6%-34.7%). The median PFS and OS were 3.1 months (95% CI: 1.5-4.3) and 6.8 months (95% CI: 4.7-10.1), respectively. A numerical difference without any statistical significance in ORR was observed in patients with different EGFR expressions (≥ 50%: 25.0% vs. < 50%: 0%, P = 0.140) or TP53 mutation abundance (< 10%: 23.8% vs. ≥ 10%: 0%, P = 0.286). Improved median PFS (3.4 vs. 1.4 months, P = 0.006) and OS (8.0 vs. 4.2 months, P = 0.027) were associated with TP53 mutation abundance of < 10%. The most common treatment-related adverse events of grade 3 or 4 (occurring in ≥ 2 patients) were hypomagnesemia [7 (23.3%)] and rash [2 (6.7%)]. No treatment-related death occurred. CONCLUSIONS SCT200 monotherapy as the second- or further-line treatment for advanced ESCC showed favorable efficacy, with an acceptable safety profile. TP53 mutation abundance might serve as a potential predictive biomarker.
Collapse
Affiliation(s)
- Ming Bai
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin’s Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China
| | - Meng Wang
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin’s Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China
| | - Ting Deng
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin’s Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China
| | - Yuxian Bai
- Department of Gastrointestinal Oncology, Harbin Medical University Cancer Hospital, Harbin 150081, China
| | - Kai Zang
- Department of Medical Oncology, Henan Cancer Hospital, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou 450008, China
| | - Zhanhui Miao
- Oncology Department, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang 453100, China
| | - Wenlin Gai
- Sinocelltech Ltd., Beijing 100176, China
| | - Liangzhi Xie
- Sinocelltech Ltd., Beijing 100176, China
- Beijing Engineering Research Center of Protein and Antibody, Beijing 100176, China
- Cell Culture Engineering Center, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100176, China
| | - Yi Ba
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin’s Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China
| |
Collapse
|
9
|
A comprehensive review on immuno-nanomedicine for breast cancer therapy: Technical challenges and troubleshooting measures. Int Immunopharmacol 2021; 103:108433. [PMID: 34922248 DOI: 10.1016/j.intimp.2021.108433] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 12/01/2021] [Accepted: 12/01/2021] [Indexed: 11/21/2022]
Abstract
Nanosized drug carriers have received a major attention in cancer therapeutics and theranostics. The immuno-nanomedicine is a combination of monoclonal antibody (mAb)/mAb-drug-nanoparticles. The immuno-nanomedicine offers a promising strategy to target cancer cells. However, the understating of nanotechnology, cancer biology, immunomedicine, and nanoparticle surface chemistry has provided a better clue to prepare the effective immuno-nanomedicine for cancer therapy. Moreover, the selection of nanoparticles type and its composition is essential for development of efficient drug delivery system (DDS) to target the cancer cell site. Immuno-nanomedicine works in the ligand-receptor binding mechanism through the interaction of mAb conjugated nanoparticles and specific antigen over expressed on target cancer cells. Therefore, the selection of specific receptors in the cancer cell and their ligand is important to prepare the active immuno-nanomedicines. Moreover, the factors such as drug loading, entrapment efficiency, size, shape, and ligand conjugation of a nanocarrier are considered as major factors for a better cancer cell, internalization, drug release, and cancer cell ablation. The target-based over-expression of antigen, mAb is engineered and conjugated with nanoparticles for successful targeting of the cancer cells without causing adverse effects to normal cells. Therefore, this review analyzed the fundamental factors in the immuno-nanomedicine for breast cancer and its technical challenges in the fabrication of the antibody alone/and drug conjugated nanoparticles.
Collapse
|
10
|
Kast J, Dutta S, Upreti VV. Panitumumab: A Review of Clinical Pharmacokinetic and Pharmacology Properties After Over a Decade of Experience in Patients with Solid Tumors. Adv Ther 2021; 38:3712-3723. [PMID: 34152568 DOI: 10.1007/s12325-021-01809-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 05/26/2021] [Indexed: 01/05/2023]
Abstract
Panitumumab is a fully human monoclonal antibody that binds to the epidermal growth factor receptor (EGFR), thereby inhibiting the growth and survival of tumors expressing EGFR. Panitumumab received approval in the USA in 2006 for the treatment of wild-type RAS (defined as wild-type in both KRAS and NRAS) metastatic colorectal cancer. Over the last 10 years, the pharmacokinetic and pharmacodynamic profile of panitumumab has been studied to further evaluate its safety, efficacy, and optimal dosing regimen. In this review, we summarize the key clinical pharmacokinetic and pharmacology data for panitumumab, and considerations for its use in special populations. Panitumumab has a nonlinear pharmacokinetic profile and its approved dosing regimen (6 mg/kg every 2 weeks) is based on body weight; dose adjustments are not needed based on sex, age, or renal or hepatic impairment. Drug interactions do not occur when panitumumab is combined with chemotherapy drugs including irinotecan, paclitaxel, and carboplatin. The level of tumor EGFR expression was found to have no effect on panitumumab pharmacokinetics or efficacy. The incidence of anti-panitumumab antibodies is low; when anti-panitumumab antibodies are produced, they do not affect the efficacy, safety, or pharmacokinetics of panitumumab. In summary, the pharmacokinetic and pharmacodynamic profile of panitumumab is well suited for standard dosing, and the approved body weight-based dosing regimen maintains efficacy and safety in the treatment of wild-type RAS metastatic colorectal cancer across a broad range of patients.
Collapse
|
11
|
Vacchelli E, Aranda F, Eggermont A, Galon J, Sautès-Fridman C, Zitvogel L, Kroemer G, Galluzzi L. Trial Watch: Tumor-targeting monoclonal antibodies in cancer therapy. Oncoimmunology 2021; 3:e27048. [PMID: 24605265 PMCID: PMC3937194 DOI: 10.4161/onci.27048] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2013] [Accepted: 11/01/2013] [Indexed: 02/06/2023] Open
Abstract
In 1997, for the first time in history, a monoclonal antibody (mAb), i.e., the chimeric anti-CD20 molecule rituximab, was approved by the US Food and Drug Administration for use in cancer patients. Since then, the panel of mAbs that are approved by international regulatory agencies for the treatment of hematopoietic and solid malignancies has not stopped to expand, nowadays encompassing a stunning amount of 15 distinct molecules. This therapeutic armamentarium includes mAbs that target tumor-associated antigens, as well as molecules that interfere with tumor-stroma interactions or exert direct immunostimulatory effects. These three classes of mAbs exert antineoplastic activity via distinct mechanisms, which may or may not involve immune effectors other than the mAbs themselves. In previous issues of OncoImmunology, we provided a brief scientific background to the use of mAbs, all types confounded, in cancer therapy, and discussed the results of recent clinical trials investigating the safety and efficacy of this approach. Here, we focus on mAbs that primarily target malignant cells or their interactions with stromal components, as opposed to mAbs that mediate antineoplastic effects by activating the immune system. In particular, we discuss relevant clinical findings that have been published during the last 13 months as well as clinical trials that have been launched in the same period to investigate the therapeutic profile of hitherto investigational tumor-targeting mAbs.
Collapse
Affiliation(s)
- Erika Vacchelli
- Gustave Roussy; Villejuif, France ; INSERM, U848; Villejuif, France ; Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Centre de Recherche des Cordeliers; Paris, France ; Université Paris-Sud/Paris XI; Paris, France
| | - Fernando Aranda
- Gustave Roussy; Villejuif, France ; INSERM, U848; Villejuif, France ; Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Centre de Recherche des Cordeliers; Paris, France
| | | | - Jérôme Galon
- Université Paris Descartes/Paris V; Sorbonne Paris Cité; Paris, France ; Université Pierre et Marie Curie/Paris VI; Paris, France ; INSERM, U872; Paris, France ; Equipe 15, Centre de Recherche des Cordeliers; Paris, France
| | - Catherine Sautès-Fridman
- Université Pierre et Marie Curie/Paris VI; Paris, France ; INSERM, U872; Paris, France ; Equipe 13, Centre de Recherche des Cordeliers; Paris, France
| | - Laurence Zitvogel
- Gustave Roussy; Villejuif, France ; INSERM, U1015; CICBT507; Villejuif, France
| | - Guido Kroemer
- Pôle de Biologie; Hôpital Européen Georges Pompidou; AP-HP; Paris, France ; Metabolomics and Cell Biology Platforms; Gustave Roussy; Villejuif, France ; INSERM, U848; Villejuif, France ; Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Centre de Recherche des Cordeliers; Paris, France ; Université Paris Descartes/Paris V; Sorbonne Paris Cité; Paris, France
| | - Lorenzo Galluzzi
- Gustave Roussy; Villejuif, France ; Université Paris Descartes/Paris V; Sorbonne Paris Cité; Paris, France ; Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Centre de Recherche des Cordeliers; Paris, France
| |
Collapse
|
12
|
Galluzzi L, Vacchelli E, Fridman WH, Galon J, Sautès-Fridman C, Tartour E, Zucman-Rossi J, Zitvogel L, Kroemer G. Trial Watch: Monoclonal antibodies in cancer therapy. Oncoimmunology 2021; 1:28-37. [PMID: 22720209 DOI: 10.4161/onci.1.1.17938] [Citation(s) in RCA: 79] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Since the advent of hybridoma technology, dating back to 1975, monoclonal antibodies have become an irreplaceable diagnostic and therapeutic tool for a wide array of human diseases. During the last 15 years, several monoclonal antibodies (mAbs) have been approved by FDA for cancer therapy. These mAbs are designed to (1) activate the immune system against tumor cells, (2) inhibit cancer cell-intrinsic signaling pathways, (3) bring toxins in the close proximity of cancer cells, or (4) interfere with the tumor-stroma interaction. More recently, major efforts have been made for the development of immunostimulatory mAbs that either enhance cancer-directed immune responses or limit tumor- (or therapy-) driven immunosuppression. Some of these antibodies, which are thought to facilitate tumor eradication by initiating or sustaining a tumor-specific immune response, have already entered clinical trials. In this Trial Watch, we will review and discuss the clinical progress of the most important mAbs that are have entered clinical trials after January 2008.
Collapse
Affiliation(s)
- Lorenzo Galluzzi
- INSERM, U848; Villejuif, France ; Institut Gustave Roussy; Villejuif, France ; Université Paris-Sud/Paris XI; Le Kremlin-Bicêtre, France
| | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Panahi Y, Mohammadzadeh AH, Behnam B, Orafai HM, Jamialahmadi T, Sahebkar A. A Review of Monoclonal Antibody-Based Treatments in Non-small Cell Lung Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1286:49-64. [PMID: 33725344 DOI: 10.1007/978-3-030-55035-6_3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Non-small cell lung cancer (NSCLC) is one of the most common types of lung cancer worldwide. It metastasizes rapidly and has a poor prognosis. The first-line treatment for most patients is a combination of chemotherapy and radiation. In many subjects, using targeted treatments alongside chemoradiation has shown a better outcome in terms of progression and quality of life for patients. These targeted treatments include small biological inhibiting molecules and monoclonal antibodies. In this review, we have assessed studies focused upon the treatment of non-small cell lung cancer. Some therapies are approved, such as bevacizumab and atezolizumab, while some are still in clinical trials, such as ficlatuzumab and ipilimumab, and others have been rejected due to inadequate disease control, such as figitumumab.
Collapse
Affiliation(s)
- Yunes Panahi
- Pharmacotherapy Department, Faculty of Pharmacy, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Amir Hossein Mohammadzadeh
- Chemical Injuries Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Behzad Behnam
- Pharmaceutics Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran.,Herbal and Traditional Medicines Research Center, Kerman University of Medical Sciences, Kerman, Iran.,Faculty of Pharmacy, Department of Pharmaceutical Biotechnology, Kerman University of Medical Sciences, Kerman, Iran
| | - Hossein M Orafai
- Faculty of Pharmacy, Department of Pharmaceutics, University of Ahl Al Bayt, Karbala, Iraq.,Faculty of Pharmacy, Department of Pharmaceutics, Al-Zahraa University, Karbala, Iraq
| | - Tannaz Jamialahmadi
- Department of Food Science and Technology, Quchan Branch, Islamic Azad University, Quchan, Iran.,Faculty of Medicine, Department of Nutrition, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amirhossein Sahebkar
- Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran. .,Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran. .,Polish Mother's Memorial Hospital Research Institute (PMMHRI), Lodz, Poland.
| |
Collapse
|
14
|
Chung TK, Lee HA, Park SI, Oh DY, Lee KW, Kim JW, Kim JH, Woo A, Lee SJ, Bang YJ, Lee H. A target-mediated drug disposition population pharmacokinetic model of GC1118, a novel anti-EGFR antibody, in patients with solid tumors. Clin Transl Sci 2021; 14:990-1001. [PMID: 33382918 PMCID: PMC8212746 DOI: 10.1111/cts.12963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 11/18/2020] [Accepted: 12/03/2020] [Indexed: 11/29/2022] Open
Abstract
Abstract GC1118 is a monoclonal antibody for epidermal growth factor receptor (EGFR) that is currently under clinical development to treat patients with solid tumors. In this study, the pharmacokinetics (PKs) of GC1118 were modeled in solid tumor patients who received a 2‐h intravenous infusion of GC1118 at 0.3, 1, 3, 5, or 4 mg/kg once‐weekly (Q1W) on days 1, 8, 15, and 22 or 8 mg/kg every other week on days 1 and 15. A target‐mediated drug disposition population PK model adequately described the concentration‐time profiles of GC1118. Monte‐Carlo simulation experiments of the PK profiles and EGFR occupancies (ROs) by GC1118 based on the final model showed that Q1W at 4 or 5 mg/kg will produce a better antitumor effect than Q2W at 8 mg/kg. Because GC1118 was safer at 4 mg/kg than 5 mg/kg in the phase I study, we suggest to test the 4 mg/kg Q1W regimen in further clinical trials with GC1118. Study Highlights WHAT IS THE CURRENT KNOWLEDGE ON THE TOPIC?
GC1118, a fully human IgG1 monoclonal antibody (mAb) for epidermal growth factor receptor (EGFR), showed a nonlinear pharmacokinetic (PK) profile in monkeys and humans. The total clearance of GC1118 decreased as the dose was increased up to 3–4 mg/kg in humans, beyond which it remained stable. The recommended phase II dose for GC1118 was 4 mg/kg intravenously infused over 2 h once weekly.
WHAT QUESTION DID THIS STUDY ADDRESS?
We developed a target‐mediated drug disposition (TMDD) population PK model that described the nonlinear PK profile of GC1118 in patients with solid tumors. We also simulated the PK profiles and receptor occupancies for different dosage regimens.
WHAT DOES THIS STUDY ADD TO OUR KNOWLEDGE?
The TMDD population PK model adequately described the nonlinear and multiphasic PK profiles of GC1118 in humans. The simulation experiment showed that once‐weekly GC1118 at 4–5 mg/kg could be more efficacious than the biweekly regimen at 8 mg/kg.
HOW MIGHT THIS CHANGE CLINICAL PHARMACOLOGY OR TRANSLATIONAL SCIENCE?
The pharmacometrics analysis could support better informed drug development decisions for GC1118, particularly for determining an optimal dosage regimen.
Collapse
Affiliation(s)
- Tae Kyu Chung
- Department of Transdisciplinary Studies, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Korea
| | - Hyun A Lee
- Department of Transdisciplinary Studies, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Korea
| | - Sang-In Park
- Department of Pharmacology, College of Medicine, Kangwon National University, Chuncheon, Korea
| | - Do-Youn Oh
- Department of Internal Medicine, Seoul National University Hospital, Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Keun-Wook Lee
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Jin Won Kim
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Jee Hyun Kim
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | | | | | - Yung-Jue Bang
- Department of Internal Medicine, Seoul National University Hospital, Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Howard Lee
- Department of Transdisciplinary Studies, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Korea.,Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Hospital, Seoul, Korea.,Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Korea.,Center for Convergence Approaches in Drug Development, Seoul National University, Seoul, Korea.,Advanced Institute of Convergence Technology, Suwon, Korea
| |
Collapse
|
15
|
Abstract
PURPOSE OF REVIEW Dysphagia is a debilitating, depressing and potentially life-threatening complication in cancer patients that is likely underreported. The purpose of this review is to critically synthesize the current knowledge regarding the impact of chemotherapeutic regimens on swallowing function. RECENT FINDINGS Those patients with cancers involving the aerodigestive tract, head and neck cancer and oesophageal cancer are at highest risk of developing dysphagia. The most common dysphagia causing toxicity of chemotherapeutic agents is mucositis/stomatitis. The use of cisplatin is correlated with increased incidence of mucositis. Similarly, the addition of melphalan is also associated with worsening mucositis and dysphagia. In some cases of oesophageal cancer, thyroid cancer, metastatic lung or breast cancer the use of chemotherapy can improve swallow function as obstructive lesions are reduced. SUMMARY There is limited literature regarding the role of chemotherapy in the development or treatment of dysphagia. Most dysphagia that occurs during cancer treatment is attributable to radiation or the synergistic effect of radiation and chemotherapy. Patients with disordered swallowing prior to treatment have the greatest risk of developing posttreatment dysphagia. Studies are needed to determine whether acute inflammation associated with oropharyngeal mucositis predisposes for late dysphagia.
Collapse
|
16
|
Clinical Pharmacokinetics and Pharmacodynamics of the Epidermal Growth Factor Receptor Inhibitor Panitumumab in the Treatment of Colorectal Cancer. Clin Pharmacokinet 2019; 57:455-473. [PMID: 28853050 PMCID: PMC5856878 DOI: 10.1007/s40262-017-0590-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Despite progress in the treatment of metastatic colorectal cancer (mCRC) in the last 15 years, it is still a condition with a relatively low 5-year survival rate. Panitumumab, a fully human monoclonal antibody directed against the epidermal growth factor receptor (EGFR), is able to prolong survival in patients with mCRC. Panitumumab is used in different lines of therapy in combination with chemotherapy, and as monotherapy for the treatment of wild-type (WT) RAS mCRC. It is administered as an intravenous infusion of 6 mg/kg every 2 weeks and has a t½ of approximately 7.5 days. Elimination takes place via two different mechanisms, and immunogenicity rates are low. Only RAS mutations have been confirmed as a negative predictor of efficacy with anti-EGFR antibodies. Panitumumab is generally well tolerated and has a manageable toxicity profile, despite a very high prevalence of dermatologic side effects. This article presents an overview of the clinical pharmacokinetics and pharmacodynamics of panitumumab, including a description of the studies that led to its approval in the different lines of therapy of mCRC.
Collapse
|
17
|
McGregor M, Price TJ. Panitumumab in the treatment of metastatic colorectal cancer, including wild-type RAS, KRAS and NRAS mCRC. Future Oncol 2018; 14:2437-2459. [PMID: 29737864 DOI: 10.2217/fon-2017-0711] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The humanized monoclonal antibody panitumumab, targeted against EGFR, plays an important role in patients with metastatic colorectal cancer. This article reviews the body of evidence for panitumumab which demonstrates significant benefits across multiple lines of therapy in those without an extended RAS mutation. The use of panitumumab with RAS mutations is not beneficial and possibly harmful. Panitumumab is well tolerated with manageable toxicities. The role of panitumumab continues to evolve as understanding of sequencing of therapies grows. There is evidence for use as maintenance therapy and conversion therapy for unresectable liver metastases. Future research is likely to focus on biomarkers for improved patient selection and the development of novel therapeutic strategies to overcome resistance.
Collapse
Affiliation(s)
- Mark McGregor
- Medical Oncology, Adelaide Oncology & Haematology, North Adelaide, Australia.,Medical Oncology, Flinders Medical Centre, Adelaide, Australia
| | - Timothy J Price
- Medical Oncology, Adelaide Oncology & Haematology, North Adelaide, Australia.,Medical Oncology, The Queen Elizabeth Hospital & University of Adelaide, Woodville, Adelaide, Australia
| |
Collapse
|
18
|
Development and validation of an ELISA to study panitumumab pharmacokinetics. Bioanalysis 2018; 10:205-214. [DOI: 10.4155/bio-2016-0292] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Aim: Panitumumab is a monoclonal antibody directed against EGFR that is approved for the treatment of metastatic colorectal cancer. To investigate its pharmacokinetics and concentration–response relationship, a validated assay is required. Results: An ELISA assay was developed and validated according to international recommendations. Six calibrators (ranging from 0.1 to 20 mg/l) plus one anchor point (50 mg/l) and three quality controls (0.45, 2 and 8 mg/l) were defined. The limit of detection, lower limit of quantification and upper limit of quantification were 0.033, 0.112 and 10 mg/l, respectively. Conclusion: This method is validated and can be used to study pharmacokinetics of panitumumab or to perform therapeutic drug monitoring.
Collapse
|
19
|
Spigel DR, Mekhail TM, Waterhouse D, Hadley T, Webb C, Burris HA, Hainsworth JD, Greco FA. First-Line Carboplatin, Pemetrexed, and Panitumumab in Patients with Advanced Non-Squamous KRAS Wild Type (WT) Non-Small-Cell Lung Cancer (NSCLC). Cancer Invest 2017; 35:541-546. [PMID: 28762849 DOI: 10.1080/07357907.2017.1344698] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
BACKGROUND We added panitumumab to standard combination chemotherapy as first-line treatment for patients with advanced KRAS WT non-squamous NSCLC. METHODS Patients received panitumumab 9 mg/kg IV, pemetrexed 500 mg/m2 IV, and carboplatin AUC = 6 IV every 21 days. After 6 cycles, maintenance therapy with panitumumab and pemetrexed was administered every 21 days until progressive disease or unacceptable toxicity. RESULTS 29 of 66 patients (44%) had objective responses. The median TTP was 6 months; median overall survival (OS) was 17 months. Panitumumab increased treatment-related toxicity, notably skin rash. CONCLUSIONS The addition of panitumumab increased toxicity, and had no discernible impact on efficacy.
Collapse
Affiliation(s)
- David R Spigel
- a Sarah Cannon Research Institute/Tennessee Oncology, PLLC , Nashville , Tennessee , USA
| | - Tarek M Mekhail
- b Florida Hospital Cancer Institute , Orlando , Florida , USA
| | - David Waterhouse
- c Oncology Hematology Care/US Oncology , Cincinnati , Ohio , USA
| | | | - Charles Webb
- e Baptist Hospital East , Louisville , Kentucky , USA
| | - Howard A Burris
- a Sarah Cannon Research Institute/Tennessee Oncology, PLLC , Nashville , Tennessee , USA
| | - John D Hainsworth
- a Sarah Cannon Research Institute/Tennessee Oncology, PLLC , Nashville , Tennessee , USA
| | - F Anthony Greco
- a Sarah Cannon Research Institute/Tennessee Oncology, PLLC , Nashville , Tennessee , USA
| |
Collapse
|
20
|
Ferris RL, Geiger JL, Trivedi S, Schmitt NC, Heron DE, Johnson JT, Kim S, Duvvuri U, Clump DA, Bauman JE, Ohr JP, Gooding WE, Argiris A. Phase II trial of post-operative radiotherapy with concurrent cisplatin plus panitumumab in patients with high-risk, resected head and neck cancer. Ann Oncol 2016; 27:2257-2262. [PMID: 27733374 DOI: 10.1093/annonc/mdw428] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2016] [Revised: 08/31/2016] [Accepted: 09/01/2016] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Treatment intensification for resected, high-risk, head and neck squamous cell carcinoma (HNSCC) is an area of active investigation with novel adjuvant regimens under study. In this trial, the epidermal growth-factor receptor (EGFR) pathway was targeted using the IgG2 monoclonal antibody panitumumab in combination with cisplatin chemoradiotherapy (CRT) in high-risk, resected HNSCC. PATIENTS AND METHODS Eligible patients included resected pathologic stage III or IVA squamous cell carcinoma of the oral cavity, larynx, hypopharynx, or human-papillomavirus (HPV)-negative oropharynx, without gross residual tumor, featuring high-risk factors (margins <1 mm, extracapsular extension, perineural or angiolymphatic invasion, or ≥2 positive lymph nodes). Postoperative treatment consisted of standard RT (60-66 Gy over 6-7 weeks) concurrent with weekly cisplatin 30 mg/m2 and weekly panitumumab 2.5 mg/kg. The primary endpoint was progression-free survival (PFS). RESULTS Forty-six patients were accrued; 44 were evaluable and were analyzed. The median follow-up for patients without recurrence was 49 months (range 12-90 months). The probability of 2-year PFS was 70% (95% CI = 58%-85%), and the probability of 2-year OS was 72% (95% CI = 60%-87%). Fourteen patients developed recurrent disease, and 13 (30%) of them died. An additional five patients died from causes other than HNSCC. Severe (grade 3 or higher) toxicities occurred in 14 patients (32%). CONCLUSIONS Intensification of adjuvant treatment adding panitumumab to cisplatin CRT is tolerable and demonstrates improved clinical outcome for high-risk, resected, HPV-negative HNSCC patients. Further targeted monoclonal antibody combinations are warranted. REGISTERED CLINICAL TRIAL NUMBER NCT00798655.
Collapse
Affiliation(s)
- R L Ferris
- Cancer Immunology Program, University of Pittsburgh Cancer Institute, Pittsburgh .,Departments of Otolaryngology, Division of Head and Neck Surgery.,Immunology
| | - J L Geiger
- Internal Medicine, Division of Hematology/Oncology, University of Pittsburgh, Pittsburgh
| | - S Trivedi
- Departments of Otolaryngology, Division of Head and Neck Surgery
| | - N C Schmitt
- Department of Otolaryngology, Johns Hopkins University, Baltimore.,Tumor Biology Section, National Institute of Deafness and Communication Disorders, National Institutes of Health, Bethesda
| | - D E Heron
- Cancer Immunology Program, University of Pittsburgh Cancer Institute, Pittsburgh.,Departments of Otolaryngology, Division of Head and Neck Surgery.,Department of Radiation Oncology, University of Pittsburgh, Pittsburgh, USA
| | - J T Johnson
- Departments of Otolaryngology, Division of Head and Neck Surgery
| | - S Kim
- Departments of Otolaryngology, Division of Head and Neck Surgery
| | - U Duvvuri
- Departments of Otolaryngology, Division of Head and Neck Surgery
| | - D A Clump
- Cancer Immunology Program, University of Pittsburgh Cancer Institute, Pittsburgh.,Department of Otolaryngology, Johns Hopkins University, Baltimore
| | - J E Bauman
- Cancer Immunology Program, University of Pittsburgh Cancer Institute, Pittsburgh.,Internal Medicine, Division of Hematology/Oncology, University of Pittsburgh, Pittsburgh
| | - J P Ohr
- Cancer Immunology Program, University of Pittsburgh Cancer Institute, Pittsburgh.,Internal Medicine, Division of Hematology/Oncology, University of Pittsburgh, Pittsburgh
| | - W E Gooding
- Cancer Immunology Program, University of Pittsburgh Cancer Institute, Pittsburgh
| | - A Argiris
- Department of Medical Oncology, Hygeia Hospital, Athens, Greece.,Department of Medical Oncology, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, USA
| |
Collapse
|
21
|
Daoud MA, Aboelnaga EM, Mohamed WM. Second-line panitumumab as a triweekly dose for patients with wild-type KRAS exon 2 metastatic colorectal cancer: a single-institution experience. Cancer Biol Med 2016; 13:136-41. [PMID: 27144068 PMCID: PMC4850122 DOI: 10.28092/j.issn.2095-3941.2015.0009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
OBJECTIVE Panitumumab administered as monotherapy in colorectal cancer (CRC) has shown response and disease stabilization rates of approximately 30%. The current study aimed to evaluate the progression-free survival (PFS) and overall survival (OS) of patients with metastatic colorectal cancer (mCRC) treated with panitumumab every 3 weeks as a second line treatment. METHODS This study is a retrospective analysis of 18 patients, aged more than 18 years, with wild-type KRAS exon 2 mCRC treated with panitumumab as a second-line single agent after progression on first-line chemotherapy. RESULTS The median number of courses received was 10 (range, 4-29), and the median duration of treatment was 30 weeks (range, 12-96 weeks). After a median follow-up period of 13 months, the median PFS was 6 months (range, 4.3-7.7 months) and the median OS was 11 months (range, 7.4-14.5 months). The median PFS was 4 months for patients with < grade 2 skin toxicity and 6 months (range, 4.5-7.5 months) for patients with ≥grade 2 skin rash (P=0.05). The median OS was 9 months (range, 6.4-11.5 months) and 14 months (range, 11.6-16.3 months) for the two groups of patients (P=0.002). CONCLUSIONS Panitumumab given every 3 weeks is effective and well tolerated in patients with advanced CRC that progressed after standard chemotherapy.
Collapse
Affiliation(s)
- Mohamed A. Daoud
- Department of Radiation Oncology, Mansoura Faculty of Medicine, King Abdullah Medical City, Mecca 24246, Saudi Arabia
| | - Engy M. Aboelnaga
- Department of Radiation Oncology, Mansoura Faculty of Medicine, King Abdullah Medical City, Mecca 24246, Saudi Arabia
| | - Wael M. Mohamed
- Department of Radiation Oncology, Mansoura Faculty of Medicine, King Abdullah Medical City, Mecca 24246, Saudi Arabia
| |
Collapse
|
22
|
Abstract
Antibody-based immunotherapy has become a standard treatment for a variety of cancers. Many well-developed antibodies disrupt signaling of various growth factor receptors for the treatment of a number of cancers by targeting surface antigens expressed on tumor cells. In recent years, a new family of antibodies is currently emerging in the clinic, which target immune cells rather than cancer cells. These immune-targeted therapies strive to augment antitumor immune responses by antagonizing immunosuppressive pathways or providing exogenous immune-activating stimuli, which have achieved dramatic results in several cancers. The future of cancer therapies is likely to combine these approaches with other treatments, including conventional therapies, to generate more effective treatments.
Collapse
Affiliation(s)
- Shengdian Wang
- CAS Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Datun Road #15, Chaoyang District, 100101, Beijing, China.
| | - Mingming Jia
- CAS Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Datun Road #15, Chaoyang District, 100101, Beijing, China
| |
Collapse
|
23
|
Patel SB, Gill D, Garrido-Laguna I. Profile of panitumumab as first-line treatment in patients with wild-type KRAS metastatic colorectal cancer. Onco Targets Ther 2015; 9:75-86. [PMID: 26770060 PMCID: PMC4706127 DOI: 10.2147/ott.s68558] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Targeted therapies against EGFR, vascular endothelial growth factor, and vascular endothelial growth factor receptor have expanded treatment options for patients with metastatic colorectal cancer (mCRC). Unfortunately, biomarkers to identify patients that are most likely to derive benefit from targeted therapies in this disease are still needed. Indeed, only RAS mutations have been identified as predictive of lack of benefit from monoclonal antibodies against EGFR in patients with mCRC. Panitumumab is a fully humanized monoclonal antibody against EGFR. In this study, we review data to support the use of panitumumab in combination with a chemotherapy backbone, in the first line setting in patients with RAS wild-type mCRC. Ongoing efforts are aimed at identifying smaller subsets of patients within the RAS wild-type group that will derive the largest benefit from anti-EGFR therapy. In the meantime, treatment with anti-EGFR therapy should be reserved for patients with RAS wild-type mCRC.
Collapse
Affiliation(s)
- Shiven B Patel
- Department of Internal Medicine, Oncology Division and Center for Investigational Therapeutics, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - David Gill
- Department of Internal Medicine, Oncology Division and Center for Investigational Therapeutics, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Ignacio Garrido-Laguna
- Department of Internal Medicine, Oncology Division and Center for Investigational Therapeutics, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| |
Collapse
|
24
|
Lo L, Patel D, Townsend AR, Price TJ. Pharmacokinetic and pharmacodynamic evaluation of panitumumab in the treatment of colorectal cancer. Expert Opin Drug Metab Toxicol 2015; 11:1907-24. [PMID: 26572750 DOI: 10.1517/17425255.2015.1112787] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
INTRODUCTION Integration of targeted therapy and additional chemotherapy options has improved median overall survival (OS) in patients with unresectable metastatic colorectal cancer (mCRC). Cetuximab and panitumumab are examples of targeted therapies, specifically against the epidermal growth factor receptor (EGFR). This review focuses on Panitumumab, a fully human IgG2 monoclonal antibody, which inhibits key oncogenic downstream cell signalling pathways. Panitumumab and cetuximab have improved tumour response rate, progression-free survival, and OS in mCRC patients in whom the RAS (Rat Sarcoma) gene is of Wild Type (WT) status. AREAS COVERED The EGFR signalling pathway and preclinical, Phase I and Phase II clinical studies on the pharmacokinetic, pharmacodynamic and safety evaluation of panitumumab are presented. Phase III studies utilising panitumumab in the first, second and third line setting in mCRC are also described. EXPERT OPINION Panitumumab exhibits excellent pharmacokinetics and pharmacodynamics by way of uncomplicated dosing, non-existent drug interactions, minimal infusion reactions and manageable side effects, making it a suitable target for combination treatments. However, innate and acquired resistances are still obstacles. To overcome this, experimented strategies are ongoing, particularly in patients with Her-2 and BRAF gene alterations. Novel biomarkers to improve patient selection and second-generation targeted antibodies are in development.
Collapse
Affiliation(s)
- Louisa Lo
- a Department of Medical Oncology , The Queen Elizabeth Hospital , Woodville , 5011 , SA , Australia
| | - Dainik Patel
- a Department of Medical Oncology , The Queen Elizabeth Hospital , Woodville , 5011 , SA , Australia
| | - Amanda R Townsend
- a Department of Medical Oncology , The Queen Elizabeth Hospital , Woodville , 5011 , SA , Australia.,b School of Medicine , University of Adelaide , Adelaide , 5000 , SA , Australia
| | - Timothy J Price
- a Department of Medical Oncology , The Queen Elizabeth Hospital , Woodville , 5011 , SA , Australia.,b School of Medicine , University of Adelaide , Adelaide , 5000 , SA , Australia
| |
Collapse
|
25
|
Sheng J, Yang YP, Zhao YY, Qin T, Hu ZH, Zhou T, Zhang YX, Hong SD, Ma YX, Zhao HY, Huang Y, Zhang L. The Efficacy of Combining EGFR Monoclonal Antibody With Chemotherapy for Patients With Advanced Nonsmall Cell Lung Cancer: A Meta-Analysis From 9 Randomized Controlled Trials. Medicine (Baltimore) 2015; 94:e1400. [PMID: 26313787 PMCID: PMC4602912 DOI: 10.1097/md.0000000000001400] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Although epidermal growth factor receptor (EGFR) monoclonal antibodies (mAbs) have been proved synergistic effect when combined with cytotoxic agents for advanced nonsmall cell lung cancer (NSCLC), the results of relevant clinical trials remain controversial. The purpose of this meta-analysis was to assess the advantage and toxicity profile of chemotherapy plus EGFR-mAbs versus chemotherapy alone for patients with NSCLC.We rigorously searched electronic databases for eligible studies reporting EGFR-mAbs combined with chemotherapy versus chemotherapy alone for patients with advanced NSCLC. The primary outcome was overall survival (OS). Pooled results were calculated using proper statistical methods.Nine phase II/III randomized controlled trials involved a total of 4949 participants were included. In general, compared with chemotherapy alone, the addition of EGFR-mAbs significantly improved OS (hazard ratio [HR] = 0.91, 95% confidence interval [CI]: 0.86-0.97, P = 0.006), progression-free survival (HR = 0.83, 95% CI: 0.87-0.98, P = 0.01), response rate (odd ratio [OR] = 1.28, 95% CI: 1.12-1.47, P = 0.0003), and disease control rate (OR = 1.17, 95% CI: 1.01-1.36, P = 0.04). Subgroup analysis showed that apparent OS benefit present in patients with squamous NSCLC (HR = 0.83, 95% CI: 0.74-0.93, P = 0.001), and those treatment-naive population (HR = 0.88, 95% CI: 0.82-0.95, P = 0.0006). Several manageable adverse events were markedly increased by EGFR-mAbs, such as acne-like rash, infusion reactions, and diarrhea. The risk for some ≥Grade 3 toxicities, such as leukopenia, febrile neutropenia, and thromboembolic events were slightly increased by the addition of EGFR-mAbs. In general, the toxicities of the combination strategy were tolerable and manageable.The addition of EGFR-mAbs to chemotherapy provided superior clinical benefit along with acceptable toxicities to patients with advanced NSCLC, especially those harboring squamous cancer and treatment-naive. Further validation in front-line investigation, proper selection of the potential benefit population by tumor histology, and development of prognostic biomarkers are warranted for future research and clinical application of EGFR-mAbs.
Collapse
Affiliation(s)
- Jin Sheng
- From the Department of Medical Oncology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, P.R. China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
You B, Chen EX. Anti-EGFR monoclonal antibodies for treatment of colorectal cancers: development of cetuximab and panitumumab. J Clin Pharmacol 2015; 52:128-55. [PMID: 21427284 DOI: 10.1177/0091270010395940] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Over the last decade, anti-epidermal growth factor receptor (EGFR) monoclonal antibodies (mAbs) have been firmly established as essential drugs for the treatment of metastatic colorectal cancer (CRC). Cetuximab and panitumumab have been approved by American and European drug agencies. This review aims at exploring the main outcomes of clinical studies performed during their clinical development, from phase I to III trials, and hence at giving a comprehensive review of the scientific rational and up-to-date evidence sustaining the use of these drugs. Many areas are still under active investigation such as administration schedules, their efficacy in comparison with bevacizumab, their role in adjuvant therapy, molecular predictors, and management of side effects.
Collapse
Affiliation(s)
- Benoit You
- Department of Medical Oncology and Hematology, Princess Margaret Hospital, University Health Network, Toronto, Ontario, Canada
| | | |
Collapse
|
27
|
Osawa M, Kudoh S, Sakai F, Endo M, Hamaguchi T, Ogino Y, Yoneoka M, Sakaguchi M, Nishimoto H, Gemma A. Clinical features and risk factors of panitumumab-induced interstitial lung disease: a postmarketing all-case surveillance study. Int J Clin Oncol 2015; 20:1063-71. [PMID: 25967287 PMCID: PMC4666285 DOI: 10.1007/s10147-015-0834-3] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Accepted: 04/14/2015] [Indexed: 12/03/2022]
Abstract
Background Drug-induced interstitial lung disease (ILD) is one of the most serious adverse reactions associated with the molecularly targeted drugs. Panitumumab has been approved for advanced or recurrent colorectal cancer. Although there were no adverse reaction reports of ILD in panitumumab monotherapy, 4 cases in combination chemotherapy were reported prior to its approval in Japan in 2010. Several studies also reported that the incidence of drug-induced ILD was higher in Japan than in other countries. The clinical features of ILD and the associated risk factors therefore need investigation. Methods We analyzed the data from 3085 unresectable, advanced or recurrent colorectal cancer patients enrolled in a postmarketing all-case surveillance study of panitumumab in Japan. ILD case reports were assessed based on the clinical and radiologic findings by a committee of external experts. Multivariate analysis using Cox’s hazard model identified the risk factors. Results ILD incidence (1.3 %) and mortality rates (51.3 %) were similar to those of patients receiving another anti-epidermal growth factor receptor (EGFR) monoclonal antibody in Japan. No specific onset timing was determined. Although panitumumab-specific ILD findings were not observed in computed tomography images or clinical practice, panitumumab can induce ILD with diffuse alveolar damage, as do the other anti-EGFR targeting drugs. A history/complication of ILD, male sex, poor general condition, and 65 years or older were identified as ILD risk factors, and no history of previous drug treatment was an apparent risk factor. Conclusion Panitumumab-induced ILD can occur at any time after initiation, and close and regular monitoring is needed.
Collapse
Affiliation(s)
- Masahiro Osawa
- Pharmacovigilance Department, Takeda Pharmaceutical Company Limited, 4-9, Hiranomachi 2-chome, Chuo-ku, Osaka, 541-0046, Japan.
| | - Shoji Kudoh
- Fukujuji Hospital, Kiyose, Japan.,Japan Anti-Tuberculosis Association, Tokyo, Japan
| | - Fumikazu Sakai
- Department of Diagnostic Radiology, Saitama International Medical Center, Saitama Medical University, Hidaka, Japan
| | - Masahiro Endo
- Division of Diagnostic Radiology, Shizuoka Cancer Center, Nagaizumi-Cho, Shizuoka, Japan
| | - Tetsuya Hamaguchi
- Department of Gastrointestinal Medical Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Yumiko Ogino
- Pharmacovigilance Department, Takeda Pharmaceutical Company Limited, 4-9, Hiranomachi 2-chome, Chuo-ku, Osaka, 541-0046, Japan
| | - Miyo Yoneoka
- Pharmacovigilance Department, Takeda Pharmaceutical Company Limited, 4-9, Hiranomachi 2-chome, Chuo-ku, Osaka, 541-0046, Japan
| | - Motonobu Sakaguchi
- Pharmacovigilance Department, Takeda Pharmaceutical Company Limited, 4-9, Hiranomachi 2-chome, Chuo-ku, Osaka, 541-0046, Japan
| | - Hiroyuki Nishimoto
- Pharmacovigilance Department, Takeda Pharmaceutical Company Limited, 4-9, Hiranomachi 2-chome, Chuo-ku, Osaka, 541-0046, Japan
| | - Akihiko Gemma
- Department of Pulmonary Medicine and Oncology, Graduate School of Medicine, Nippon Medical School, Tokyo, Japan
| |
Collapse
|
28
|
Mizugaki H, Yamamoto N, Fujiwara Y, Nokihara H, Yamada Y, Tamura T. Current Status of Single-Agent Phase I Trials in Japan: Toward Globalization. J Clin Oncol 2015; 33:2051-61. [PMID: 25918301 DOI: 10.1200/jco.2014.58.4953] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
PURPOSE In Japan, phase I trials, except first-in-human trials, are usually initiated from approximately 50% of the maximum-tolerated dose (MTD) or maximum administered dose (MAD) determined during the initial phase I trials in North America and Europe (the West). However, the key findings of phase I trials in Japan and the West, such as dose-limiting toxicity (DLT) profiles and MTD or MAD levels, have not been compared. PATIENTS AND METHODS We retrospectively analyzed data for patients enrolled onto single-agent phase I trials at the National Cancer Center Hospital between 1995 and 2012. DLT profiles, MTDs, and MADs of single-agent phase I trials in Japan were compared with those from trials in the West that were obtained from the literature. RESULTS A total of 777 patients were enrolled onto 54 single-agent phase I trials, including five first-in-human trials. DLTs were observed in 11.1% of the patients. Importantly, 66.4% of the DLTs were observed within a dose range (80% to 120%) similar to those reported for the trials in the West. The majority of MTDs or MADs could be considered similar between patients, and 80.3% of the drugs had similar MTDs or MADs in the West. CONCLUSION The toxicity profiles of single-agent phase I agents determined from trials conducted in Japan were comparable to those obtained from trials in the West. We believe that phase I trials in Japan could be conducted over timelines similar to those in the West, allowing for global or parallel phase I clinical trials.
Collapse
|
29
|
Vacchelli E, Pol J, Bloy N, Eggermont A, Cremer I, Fridman WH, Galon J, Marabelle A, Kohrt H, Zitvogel L, Kroemer G, Galluzzi L. Trial watch: Tumor-targeting monoclonal antibodies for oncological indications. Oncoimmunology 2015; 4:e985940. [PMID: 25949870 DOI: 10.4161/2162402x.2014.985940] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2014] [Accepted: 05/11/2014] [Indexed: 12/31/2022] Open
Abstract
An expanding panel of monoclonal antibodies (mAbs) that specifically target malignant cells or intercept trophic factors delivered by the tumor stroma is now available for cancer therapy. These mAbs can exert direct antiproliferative/cytotoxic effects as they inhibit pro-survival signal transduction cascades or activate lethal receptors at the plasma membrane of cancer cells, they can opsonize neoplastic cells to initiate a tumor-targeting immune response, or they can be harnessed to specifically deliver toxins or radionuclides to transformed cells. As an indication of the success of this immunotherapeutic paradigm, international regulatory agencies approve new tumor-targeting mAbs for use in cancer patients every year. Moreover, the list of indications for previously licensed molecules is frequently expanded to other neoplastic disorders as the results of large, randomized clinical trials become available. Here, we discuss recent advances in the preclinical and clinical development of tumor-targeting mAbs for oncological indications.
Collapse
Affiliation(s)
- Erika Vacchelli
- Gustave Roussy Cancer Campus ; Villejuif, France ; INSERM; U1138 ; Paris, France ; Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Centre de Recherche des Cordeliers ; Paris, France
| | - Jonathan Pol
- Gustave Roussy Cancer Campus ; Villejuif, France ; INSERM; U1138 ; Paris, France ; Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Centre de Recherche des Cordeliers ; Paris, France
| | - Norma Bloy
- Gustave Roussy Cancer Campus ; Villejuif, France ; INSERM; U1138 ; Paris, France ; Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Centre de Recherche des Cordeliers ; Paris, France
| | | | - Isabelle Cremer
- INSERM; U1138 ; Paris, France ; Equipe 13; Centre de Recherche des Cordeliers ; Paris, France ; Université Pierre et Marie Curie/Paris VI ; Paris, France
| | - Wolf Hervé Fridman
- INSERM; U1138 ; Paris, France ; Equipe 13; Centre de Recherche des Cordeliers ; Paris, France ; Université Pierre et Marie Curie/Paris VI ; Paris, France
| | - Jérôme Galon
- INSERM; U1138 ; Paris, France ; Université Pierre et Marie Curie/Paris VI ; Paris, France ; Laboratory of Integrative Cancer Immunology; Centre de Recherche des Cordeliers ; Paris, France ; Université Paris Descartes/Paris V; Sorbonne Paris Cité ; Paris, France
| | - Aurélien Marabelle
- Gustave Roussy Cancer Campus ; Villejuif, France ; INSERM ; U1015 , Villejuif, France
| | - Holbrook Kohrt
- Department of Medicine; Division of Oncology; Stanford University ; Stanford, CA, USA
| | - Laurence Zitvogel
- Gustave Roussy Cancer Campus ; Villejuif, France ; INSERM ; U1015 , Villejuif, France
| | - Guido Kroemer
- INSERM; U1138 ; Paris, France ; Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Centre de Recherche des Cordeliers ; Paris, France ; Université Paris Descartes/Paris V; Sorbonne Paris Cité ; Paris, France ; Pôle de Biologie; Hôpital Européen Georges Pompidou ; AP-HP ; Paris, France ; Metabolomics and Cell Biology Platforms; Gustave Roussy Cancer Campus ; Villejuif, France
| | - Lorenzo Galluzzi
- Gustave Roussy Cancer Campus ; Villejuif, France ; INSERM; U1138 ; Paris, France ; Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Centre de Recherche des Cordeliers ; Paris, France ; Université Paris Descartes/Paris V; Sorbonne Paris Cité ; Paris, France
| |
Collapse
|
30
|
Galluzzi L, Vacchelli E, Pedro JMBS, Buqué A, Senovilla L, Baracco EE, Bloy N, Castoldi F, Abastado JP, Agostinis P, Apte RN, Aranda F, Ayyoub M, Beckhove P, Blay JY, Bracci L, Caignard A, Castelli C, Cavallo F, Celis E, Cerundolo V, Clayton A, Colombo MP, Coussens L, Dhodapkar MV, Eggermont AM, Fearon DT, Fridman WH, Fučíková J, Gabrilovich DI, Galon J, Garg A, Ghiringhelli F, Giaccone G, Gilboa E, Gnjatic S, Hoos A, Hosmalin A, Jäger D, Kalinski P, Kärre K, Kepp O, Kiessling R, Kirkwood JM, Klein E, Knuth A, Lewis CE, Liblau R, Lotze MT, Lugli E, Mach JP, Mattei F, Mavilio D, Melero I, Melief CJ, Mittendorf EA, Moretta L, Odunsi A, Okada H, Palucka AK, Peter ME, Pienta KJ, Porgador A, Prendergast GC, Rabinovich GA, Restifo NP, Rizvi N, Sautès-Fridman C, Schreiber H, Seliger B, Shiku H, Silva-Santos B, Smyth MJ, Speiser DE, Spisek R, Srivastava PK, Talmadge JE, Tartour E, Van Der Burg SH, Van Den Eynde BJ, Vile R, Wagner H, Weber JS, Whiteside TL, Wolchok JD, Zitvogel L, Zou W, Kroemer G. Classification of current anticancer immunotherapies. Oncotarget 2014; 5:12472-508. [PMID: 25537519 PMCID: PMC4350348 DOI: 10.18632/oncotarget.2998] [Citation(s) in RCA: 319] [Impact Index Per Article: 31.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2014] [Accepted: 12/15/2014] [Indexed: 11/25/2022] Open
Abstract
During the past decades, anticancer immunotherapy has evolved from a promising therapeutic option to a robust clinical reality. Many immunotherapeutic regimens are now approved by the US Food and Drug Administration and the European Medicines Agency for use in cancer patients, and many others are being investigated as standalone therapeutic interventions or combined with conventional treatments in clinical studies. Immunotherapies may be subdivided into "passive" and "active" based on their ability to engage the host immune system against cancer. Since the anticancer activity of most passive immunotherapeutics (including tumor-targeting monoclonal antibodies) also relies on the host immune system, this classification does not properly reflect the complexity of the drug-host-tumor interaction. Alternatively, anticancer immunotherapeutics can be classified according to their antigen specificity. While some immunotherapies specifically target one (or a few) defined tumor-associated antigen(s), others operate in a relatively non-specific manner and boost natural or therapy-elicited anticancer immune responses of unknown and often broad specificity. Here, we propose a critical, integrated classification of anticancer immunotherapies and discuss the clinical relevance of these approaches.
Collapse
Affiliation(s)
- Lorenzo Galluzzi
- Equipe 11 labellisée pas la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France
- INSERM, U1138, Paris, France
- Gustave Roussy Cancer Campus, Villejuif, France
- Université Paris Descartes/Paris V, Sorbonne Paris Cité, Paris, France
| | - Erika Vacchelli
- Equipe 11 labellisée pas la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France
- INSERM, U1138, Paris, France
- Gustave Roussy Cancer Campus, Villejuif, France
| | - José-Manuel Bravo-San Pedro
- Equipe 11 labellisée pas la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France
- INSERM, U1138, Paris, France
- Gustave Roussy Cancer Campus, Villejuif, France
| | - Aitziber Buqué
- Equipe 11 labellisée pas la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France
- INSERM, U1138, Paris, France
- Gustave Roussy Cancer Campus, Villejuif, France
| | - Laura Senovilla
- Equipe 11 labellisée pas la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France
- INSERM, U1138, Paris, France
- Gustave Roussy Cancer Campus, Villejuif, France
| | - Elisa Elena Baracco
- Equipe 11 labellisée pas la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France
- INSERM, U1138, Paris, France
- Gustave Roussy Cancer Campus, Villejuif, France
- Faculté de Medicine, Université Paris Sud/Paris XI, Le Kremlin-Bicêtre, France
| | - Norma Bloy
- Equipe 11 labellisée pas la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France
- INSERM, U1138, Paris, France
- Gustave Roussy Cancer Campus, Villejuif, France
- Faculté de Medicine, Université Paris Sud/Paris XI, Le Kremlin-Bicêtre, France
| | - Francesca Castoldi
- Equipe 11 labellisée pas la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France
- INSERM, U1138, Paris, France
- Gustave Roussy Cancer Campus, Villejuif, France
- Faculté de Medicine, Université Paris Sud/Paris XI, Le Kremlin-Bicêtre, France
- Sotio a.c., Prague, Czech Republic
| | - Jean-Pierre Abastado
- Pole d'innovation thérapeutique en oncologie, Institut de Recherches Internationales Servier, Suresnes, France
| | - Patrizia Agostinis
- Cell Death Research and Therapy (CDRT) Laboratory, Dept. of Cellular and Molecular Medicine, University of Leuven, Leuven, Belgium
| | - Ron N. Apte
- The Shraga Segal Dept. of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Fernando Aranda
- Equipe 11 labellisée pas la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France
- INSERM, U1138, Paris, France
- Gustave Roussy Cancer Campus, Villejuif, France
- Group of Immune receptors of the Innate and Adaptive System, Institut d'Investigacions Biomédiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Maha Ayyoub
- INSERM, U1102, Saint Herblain, France
- Institut de Cancérologie de l'Ouest, Saint Herblain, France
| | - Philipp Beckhove
- Translational Immunology Division, German Cancer Research Center, Heidelberg, Germany
| | - Jean-Yves Blay
- Equipe 11, Centre Léon Bérard (CLR), Lyon, France
- Centre de Recherche en Cancérologie de Lyon (CRCL), Lyon, France
| | - Laura Bracci
- Dept. of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Anne Caignard
- INSERM, U1160, Paris, France
- Groupe Hospitalier Saint Louis-Lariboisière - F. Vidal, Paris, France
| | - Chiara Castelli
- Unit of Immunotherapy of Human Tumors, Dept. of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale Tumori, Milano, Italy
| | - Federica Cavallo
- Molecular Biotechnology Center, Dept. of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Estaban Celis
- Cancer Immunology, Inflammation and Tolerance Program, Georgia Regents University Cancer Center, Augusta, GA, USA
| | - Vincenzo Cerundolo
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Aled Clayton
- Institute of Cancer & Genetics, School of Medicine, Cardiff University, Cardiff, UK
- Velindre Cancer Centre, Cardiff, UK
| | - Mario P. Colombo
- Unit of Immunotherapy of Human Tumors, Dept. of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale Tumori, Milano, Italy
| | - Lisa Coussens
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
| | - Madhav V. Dhodapkar
- Sect. of Hematology and Immunobiology, Yale Cancer Center, Yale University, New Haven, CT, USA
| | | | | | - Wolf H. Fridman
- INSERM, U1138, Paris, France
- Université Paris Descartes/Paris V, Sorbonne Paris Cité, Paris, France
- Université Pierre et Marie Curie/Paris VI, Paris, France
- Equipe 13, Centre de Recherche des Cordeliers, Paris, France
| | - Jitka Fučíková
- Sotio a.c., Prague, Czech Republic
- Dept. of Immunology, 2nd Faculty of Medicine and University Hospital Motol, Charles University, Prague, Czech Republic
| | - Dmitry I. Gabrilovich
- Dept. of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jérôme Galon
- INSERM, U1138, Paris, France
- Université Paris Descartes/Paris V, Sorbonne Paris Cité, Paris, France
- Université Pierre et Marie Curie/Paris VI, Paris, France
- Laboratory of Integrative Cancer Immunology, Centre de Recherche des Cordeliers, Paris, France
| | - Abhishek Garg
- Cell Death Research and Therapy (CDRT) Laboratory, Dept. of Cellular and Molecular Medicine, University of Leuven, Leuven, Belgium
| | - François Ghiringhelli
- INSERM, UMR866, Dijon, France
- Centre Georges François Leclerc, Dijon, France
- Université de Bourgogne, Dijon, France
| | - Giuseppe Giaccone
- Center for Cancer Research, National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD, USA
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, USA
| | - Eli Gilboa
- Dept. of Microbiology and Immunology, Sylvester Comprehensive Cancer Center, University of Miami, Miller School of Medicine, Miami, FL, USA
| | - Sacha Gnjatic
- Sect. of Hematology/Oncology, Immunology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Axel Hoos
- Glaxo Smith Kline, Cancer Immunotherapy Consortium, Collegeville, PA, USA
| | - Anne Hosmalin
- Université Paris Descartes/Paris V, Sorbonne Paris Cité, Paris, France
- INSERM, U1016, Paris, France
- CNRS, UMR8104, Paris, France
- Hôpital Cochin, AP-HP, Paris, France
| | - Dirk Jäger
- National Center for Tumor Diseases, University Medical Center Heidelberg, Heidelberg, Germany
| | - Pawel Kalinski
- Dept. of Surgery, University of Pittsburgh, Pittsburgh, PA, USA
- University of Pittsburgh Cancer Institute, Hillman Cancer Center, Pittsburgh, PA, USA
- Dept. of Immunology and Infectious Diseases and Microbiology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Klas Kärre
- Dept. of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm, Sweden
| | - Oliver Kepp
- Equipe 11 labellisée pas la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France
- INSERM, U1138, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
| | - Rolf Kiessling
- Dept. of Oncology, Karolinska Institute Hospital, Stockholm, Sweden
| | - John M. Kirkwood
- University of Pittsburgh Cancer Institute Laboratory, Pittsburgh, PA, USA
| | - Eva Klein
- Dept. of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm, Sweden
| | - Alexander Knuth
- National Center for Cancer Care and Research, Hamad Medical Corporation, Doha, Qatar
| | - Claire E. Lewis
- Academic Unit of Inflammation and Tumour Targeting, Dept. of Oncology, University of Sheffield Medical School, Sheffield, UK
| | - Roland Liblau
- INSERM, UMR1043, Toulouse, France
- CNRS, UMR5282, Toulouse, France
- Laboratoire d'Immunologie, CHU Toulouse, Université Toulouse II, Toulouse, France
| | - Michael T. Lotze
- Dept. of Surgery, University of Pittsburgh, Pittsburgh, PA, USA
- University of Pittsburgh Cancer Institute, Hillman Cancer Center, Pittsburgh, PA, USA
| | - Enrico Lugli
- Unit of Clinical and Experimental Immunology, Humanitas Clinical and Research Institute, Rozzano, Italy
| | - Jean-Pierre Mach
- Dept. of Biochemistry, University of Lausanne, Epalinges, Switzerland
| | - Fabrizio Mattei
- Dept. of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Domenico Mavilio
- Unit of Clinical and Experimental Immunology, Humanitas Clinical and Research Institute, Rozzano, Italy
- Dept. of Medical Biotechnologies and Translational Medicine, University of Milan, Rozzano, Italy
| | - Ignacio Melero
- Dept. of Immunology, Centro de Investigación Médica Aplicada (CIMA), Universidad de Navarra, Pamplona, Spain
- Dept. of Oncology, Clínica Universidad de Navarra, Pamplona, Spain
| | - Cornelis J. Melief
- ISA Therapeutics, Leiden, The Netherlands
- Dept. of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, The Netherlands
| | - Elizabeth A. Mittendorf
- Research Dept. of Surgical Oncology, The University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | | | - Adekunke Odunsi
- Center for Immunotherapy, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Hideho Okada
- Dept. of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
| | | | - Marcus E. Peter
- Div. of Hematology/Oncology, Northwestern University, Feinberg School of Medicine, Chicago, IL, USA
| | - Kenneth J. Pienta
- The James Buchanan Brady Urological Institute, The Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | - Angel Porgador
- The Shraga Segal Dept. of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - George C. Prendergast
- Lankenau Institute for Medical Research, Wynnewood, PA, USA
- Dept. of Pathology, Anatomy and Cell Biology, Sidney Kimmel Medical College, Philadelphia, PA, USA
- Cell Biology and Signaling Program, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Gabriel A. Rabinovich
- Laboratorio de Inmunopatología, Instituto de Biología y Medicina Experimental (IBYME), Buenos Aires, Argentina
| | - Nicholas P. Restifo
- National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Naiyer Rizvi
- Memorial Sloan Kettering Cancer Center (MSKCC), New York, NY, USA
| | - Catherine Sautès-Fridman
- INSERM, U1138, Paris, France
- Université Paris Descartes/Paris V, Sorbonne Paris Cité, Paris, France
- Université Pierre et Marie Curie/Paris VI, Paris, France
- Equipe 13, Centre de Recherche des Cordeliers, Paris, France
| | - Hans Schreiber
- Dept. of Pathology, The Cancer Research Center, The University of Chicago, Chicago, IL, USA
| | - Barbara Seliger
- Institute of Medical Immunology, Martin Luther University Halle-Wittenberg, Halle, Germany
| | - Hiroshi Shiku
- Dept. of Immuno-GeneTherapy, Mie University Graduate School of Medicine, Tsu, Japan
| | - Bruno Silva-Santos
- Instituto de Medicina Molecular, Universidade de Lisboa, Lisboa, Portugal
| | - Mark J. Smyth
- Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
- School of Medicine, University of Queensland, Herston, Queensland, Australia
| | - Daniel E. Speiser
- Dept. of Oncology, University of Lausanne, Lausanne, Switzerland
- Ludwig Cancer Research Center, Lausanne, Switzerland
| | - Radek Spisek
- Sotio a.c., Prague, Czech Republic
- Dept. of Immunology, 2nd Faculty of Medicine and University Hospital Motol, Charles University, Prague, Czech Republic
| | - Pramod K. Srivastava
- Dept. of Immunology, University of Connecticut School of Medicine, Farmington, CT, USA
- Carole and Ray Neag Comprehensive Cancer Center, Farmington, CT, USA
| | - James E. Talmadge
- Laboratory of Transplantation Immunology, Dept. of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Eric Tartour
- Université Paris Descartes/Paris V, Sorbonne Paris Cité, Paris, France
- INSERM, U970, Paris, France
- Paris-Cardiovascular Research Center (PARCC), Paris, France
- Service d'Immunologie Biologique, Hôpital Européen Georges Pompidou (HEGP), AP-HP, Paris, France
| | | | - Benoît J. Van Den Eynde
- Ludwig Institute for Cancer Research, Brussels, Belgium
- de Duve Institute, Brussels, Belgium
- Université Catholique de Louvain, Brussels, Belgium
| | - Richard Vile
- Dept. of Molecular Medicine and Immunology, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Hermann Wagner
- Institute of Medical Microbiology, Immunology and Hygiene, Technical University Munich, Munich, Germany
| | - Jeffrey S. Weber
- Donald A. Adam Comprehensive Melanoma Research Center, Moffitt Cancer Center, Tampa, FL, USA
| | - Theresa L. Whiteside
- University of Pittsburgh Cancer Institute, Hillman Cancer Center, Pittsburgh, PA, USA
- University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Jedd D. Wolchok
- Dept. of Medicine and Ludwig Center, Memorial Sloan Kettering Cancer Center (MSKCC), New York, NY, USA
- Weill Cornell Medical College, New York, NY, USA
| | - Laurence Zitvogel
- Gustave Roussy Cancer Campus, Villejuif, France
- INSERM, U1015, Villejuif, France
- Centre d'Investigation Clinique Biothérapie 507 (CICBT507), Gustave Roussy Cancer Campus, Villejuif, France
| | - Weiping Zou
- University of Michigan, School of Medicine, Ann Arbor, MI, USA
| | - Guido Kroemer
- Equipe 11 labellisée pas la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France
- INSERM, U1138, Paris, France
- Université Paris Descartes/Paris V, Sorbonne Paris Cité, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
- Pôle de Biologie, Hôpital Européen Georges Pompidou (HEGP), AP-HP, Paris, France
| |
Collapse
|
31
|
Vacchelli E, Eggermont A, Galon J, Sautès-Fridman C, Zitvogel L, Kroemer G, Galluzzi L. Trial watch: Monoclonal antibodies in cancer therapy. Oncoimmunology 2014; 2:e22789. [PMID: 23482847 PMCID: PMC3583934 DOI: 10.4161/onci.22789] [Citation(s) in RCA: 82] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
During the past 20 years, dozens-if not hundreds-of monoclonal antibodies have been developed and characterized for their capacity to mediate antineoplastic effects, either as they activate/enhance tumor-specific immune responses, either as they interrupt cancer cell-intrinsic signal transduction cascades, either as they specifically delivery toxins to malignant cells or as they block the tumor-stroma interaction. Such an intense research effort has lead to the approval by FDA of no less than 14 distinct molecules for use in humans affected by hematological or solid malignancies. In the inaugural issue of OncoImmunology, we briefly described the scientific rationale behind the use of monoclonal antibodies in cancer therapy and discussed recent, ongoing clinical studies investigating the safety and efficacy of this approach in patients. Here, we summarize the latest developments in this exciting area of clinical research, focusing on high impact studies that have been published during the last 15 months and clinical trials launched in the same period to investigate the therapeutic profile of promising, yet hitherto investigational, monoclonal antibodies.
Collapse
Affiliation(s)
- Erika Vacchelli
- Institut Gustave Roussy; Villejuif, France ; Université Paris-Sud/Paris XI; Le Kremlin-Bicêtre, France ; INSERM; U848; Villejuif, France
| | | | | | | | | | | | | |
Collapse
|
32
|
Boyle AJ, Cao PJ, Hedley DW, Sidhu SS, Winnik MA, Reilly RM. MicroPET/CT imaging of patient-derived pancreatic cancer xenografts implanted subcutaneously or orthotopically in NOD-scid mice using (64)Cu-NOTA-panitumumab F(ab')2 fragments. Nucl Med Biol 2014; 42:71-7. [PMID: 25456837 DOI: 10.1016/j.nucmedbio.2014.10.009] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2014] [Revised: 10/12/2014] [Accepted: 10/15/2014] [Indexed: 10/24/2022]
Abstract
INTRODUCTION Our objective was to study microPET/CT imaging of patient-derived pancreatic cancer xenografts in NOD-scid mice using F(ab')2 fragments of the fully-human anti-EGFR monoclonal antibody, panitumumab (Vectibix) labeled with (64)Cu. More than 90% of pancreatic cancers are EGFR-positive. METHODS F(ab')2 fragments were produced by proteolytic digestion of panitumumab IgG or non-specific human IgG, purified by ultrafiltration then modified with NOTA chelators for complexing (64)Cu. Panitumumab IgG and Fab fragments were similarly labeled with (64)Cu. EGFR immunoreactivity was determined in competition and direct (saturation) cell binding assays. The biodistribution of (64)Cu-labeled panitumumab IgG, F(ab')2 and Fab was compared in non-tumor-bearing Balb/c mice. MicroPET/CT and biodistribution studies were performed in NOD-scid mice engrafted subcutaneously (s.c.) or orthotopically with patient-derived OCIP23 pancreatic tumors, or in NOD-scid with s.c. PANC-1 human pancreatic cancer xenografts. RESULTS Panitumumab F(ab')2 fragments were produced in high purity (>90%), derivitized with 3.2±0.7 NOTA/F(ab')2, and labeled with (64)Cu (0.3-3.6MBq/μg). The binding of (64)Cu-NOTA-panitumumab F(ab')2 to OCIP23 or PANC-1 cells was decreased significantly by an excess of panitumumab IgG. The Kd for binding of (64)Cu-NOTA-panitumumab F(ab')2 to EGFR on PANC-1 cells was 0.14±0.05nmol/L. F(ab')2 fragments exhibited more suitable normal tissue distribution for tumor imaging with (64)Cu than panitumumab IgG or Fab. Tumor uptake at 48h post injection (p.i.) of (64)Cu-NOTA-panitumumab F(ab')2 was 12.0±0.9% injected dose/g (ID/g) in s.c. and 11.8±0.9% ID/g in orthotopic OCIP23 tumors vs. 6.1±1.1% ID/g in s.c. PANC-1 xenografts. Tumor/Blood (T/B) ratios were 5:1 to 9:1 for OCIP23 and 2.4:1 for PANC-1 tumors. Tumor uptake of (64)Cu-NOTA-non-specific F(ab')2 in OCIP23 xenografts was 5-fold lower than (64)Cu-panitumumab F(ab')2. All tumor xenografts were clearly imaged by microPET/CT at 24 or 48h p.i. of (64)Cu-NOTA-panitumumab F(ab')2. CONCLUSIONS (64)Cu-panitumumab F(ab')2 fragments bound with high affinity to EGFR on pancreatic cancer cells in vitro and localized specifically in patient-derived pancreatic cancer xenografts in mice in vivo, allowing tumor visualization by microPET/CT at 24 or 48h p.i.
Collapse
Affiliation(s)
- Amanda J Boyle
- Department of Pharmaceutical Sciences, University of Toronto, Toronto, ON, Canada
| | - Ping-Jiang Cao
- Ontario Cancer Institute/Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - David W Hedley
- Ontario Cancer Institute/Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Sachdev S Sidhu
- Banting and Best Department of Medical Research, Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON, Canada
| | | | - Raymond M Reilly
- Department of Pharmaceutical Sciences, University of Toronto, Toronto, ON, Canada; Department of Medical Imaging, University of Toronto, Toronto, ON, Canada; Toronto General Research Institute, University Health Network, Toronto, ON, Canada.
| |
Collapse
|
33
|
Ochiai T, Umeki M, Miyake H, Iida T, Okumura M, Ohno K, Sakamoto M, Miyoshi N, Takahashi M, Tsumura H, Tokunaga Y, Naitou H, Fukui T. Impact of 5-fluorouracil metabolizing enzymes on chemotherapy in patients with resectable colorectal cancer. Oncol Rep 2014; 32:887-92. [PMID: 24994673 PMCID: PMC4121408 DOI: 10.3892/or.2014.3299] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2014] [Accepted: 05/13/2014] [Indexed: 01/05/2023] Open
Abstract
Although 5-fluorouracil (5-FU) is an important drug for colorectal cancer (CRC) treatment, no useful biomarker is currently available to predict treatment response. Since 5-FU is converted into active or inactive forms by orotate phosphoribosyltransferase (OPRT) or dihydropyrimidine dehydrogenase (DPD), a correlation between these enzymes and response to 5-FU has been suggested. However, such a correlation has not been investigated prospectively. Therefore, in the present study, we aimed to prospectively evaluate whether OPRT and DPD were predictive factors of the response to 5-FU treatment in patients with resectable CRC. The present investigation was designed as a multicenter prospective cohort study. OPRT and DPD activities were assessed in biopsy samples, obtained surgically from patients with resectable CRC. The OPRT/DPD ratio was calculated and the cut-off values for this ratio were determined for 5-year disease-free survival (DFS) and overall survival (OS). Patients were treated with 5-FU/leucovorin (LV) regimens and oral 5-FU. The endpoint of this study was the correlation between the OPRT/DPD ratio and 5-year DFS and OS. The cut-off value for the OPRT/DPD ratio was determined by using the maximum χ2 statistic method against 5-year DFS and OS. Sixty-eight patients were enrolled from July 2003 to May 2005. The median follow-up period was 1925 days. The OPRT/DPD ratio cut-off values for 5-year DFS and OS were 0.015 and 0.013, respectively. During the 5-year DFS and OS periods, patients with higher cut-off values had a better prognosis than those with lower ratios (P=0.03 and 0.02, respectively). In conclusion, our results suggest that the OPRT/DPD ratio could be a predictive factor for response to 5-FU/LV adjuvant chemotherapy.
Collapse
Affiliation(s)
- Takumi Ochiai
- Department of Surgery, Tobu Chiiki Hospital, Tokyo Metropolitan Health and Medical Treatment Corporation, Tokyo 125-8512, Japan
| | - Masahiko Umeki
- Department of Surgery, Hyogo Prefectural Awaji Medical Center, Hyogo 656-0021, Japan
| | - Hiroshi Miyake
- Department of Surgery, Kasukabe Municipal Hospital, Saitama 344-0067, Japan
| | - Tatsumi Iida
- Department of Surgery, Nishimino Kosei Hospital, Gifu 503-131, Japan
| | - Minoru Okumura
- Department of Surgery, Hitachi General Hospital, Ibaragi 317-0077, Japan
| | - Kazuhide Ohno
- Department of Surgery, Matsudo City Hospital, Chiba 271-0064, Japan
| | - Masashi Sakamoto
- Department of Surgery, Mitsui Memorial Hospital, Tokyo 101-8643, Japan
| | - Nobukazu Miyoshi
- Department of Surgery, Kure Kyosai Hospital, Hiroshima 737-0811, Japan
| | - Masahiko Takahashi
- Department of Surgery, Hokkaido P.W.F.A.C Asahikawa-Kosei General Hospital, Hokkaido 078-8211, Japan
| | - Hidenori Tsumura
- Department of Surgery, Koshigaya Municipal Hospital, Saitama 343-0023, Japan
| | - Yukihiko Tokunaga
- Department of Surgery, Japanpost Osaka-kita Teishin Hospital, Osaka 530-0016, Japan
| | - Haruhiko Naitou
- Department of Surgery, Hokkaido Cancer Center, Hokkaido 003-0804, Japan
| | - Takuji Fukui
- Department of Surgery, Kariya Toyota General Hospital, Aichi 448-8505, Japan
| |
Collapse
|
34
|
Stremitzer S, Sebio A, Stintzing S, Lenz HJ. Panitumumab safety for treating colorectal cancer. Expert Opin Drug Saf 2014; 13:843-51. [PMID: 24766434 DOI: 10.1517/14740338.2014.915024] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Panitumumab is a human IgG2 mAb against the EGFR, inhibiting tumor cell proliferation, survival and angiogenesis. It has demonstrated clinical efficacy in metastatic colorectal cancer (CRC) in combination with chemotherapy in first- and second-line settings and as monotherapy in third-line setting. Recently, mutations in the RAS genes have been shown to be predictive of lack of efficacy, panitumumab should be restricted to patients with RAS wild-type (wt) tumors. AREAS COVERED This review focuses on main efficacy results of panitumumab in metastatic CRC in first-, second- and third-line settings in combination with chemotherapy or as monotherapy. Additionally, we have covered safety aspects of this agent in these indications, especially in K-RAS and all RAS wt patients. These safety aspects refer to the most common toxicities (i.e., acne-like skin rash, diarrhea and hypomagnesaemia). EXPERT OPINION Panitumumab adds to the armamentarium of effective agents in the treatment of metastatic CRC. Due to its human origin, panitumumab is a well-tolerated agent with low rates of infusional reactions. Skin toxicity is frequent and should be pre-emptively treated. Other common toxicities related to panitumumab treatment, such as diarrhea and hypomagnesaemia, should be closely monitored to ensure early treatment or substitution.
Collapse
Affiliation(s)
- Stefan Stremitzer
- University of Southern California, Keck School of Medicine, Norris Comprehensive Cancer Center, Division of Medical Oncology , 1441 Eastlake Avenue, Los Angeles, CA, 90033 , USA +1 323 865 3967 ; +1 323 865 0061 ;
| | | | | | | |
Collapse
|
35
|
Hansen TF, Andersen RF, Pallisgaard N, Spindler KLG, Pløen J, Keldsen N, Lindebjerg J, Sørensen FB, Jakobsen A. A 3-weekly schedule of irinotecan and panitumumab for wild-type KRAS metastatic colorectal cancer. COLORECTAL CANCER 2014. [DOI: 10.2217/crc.13.85] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
SUMMARY Aim: We investigated the combination of irinotecan and panitumumab as a 3-weekly schedule in patients with wild-type KRAS metastatic colorectal cancer, who had progressed after standard chemotherapy. Material & methods: Patients received concomitant irinotecan (350 mg/m2) and panitumumab (9 mg/kg) once every 3 weeks. The primary end point was response rate. Secondary end points included progression-free survival (PFS), overall survival (OS) and translational research. Results: Inclusion was stopped early owing to lack of efficacy (n = 31). The response rate was 16%, median PFS and OS was 2.0 months (95% CI: 1.9–4.0) and 7.8 months (95% CI: 4.6–8.8), respectively. The most commonly encountered adverse event was skin rash (84% any grade). Pretreatment cell-free DNA levels were significantly related to disease control (p = 0.04), PFS (p = 0.04) and OS (p = 0.002), respectively. Conclusion: The present treatment regimen was less effective than expected and is not recommended. The clinical importance of cell-free DNA deserves further research.
Collapse
Affiliation(s)
- Torben Frøstrup Hansen
- Department of Oncology, Vejle Hospital part of Lillebaelt Hospital, Vejle, Denmark
- Institute of Regional Health Research, University of Southern Denmark, Odense, Denmark
| | | | - Niels Pallisgaard
- Department of Biochemistry, Vejle Hospital part of Lillebaelt Hospital, Vejle, Denmark
| | - Karen-Lise Garm Spindler
- Department of Oncology, Vejle Hospital part of Lillebaelt Hospital, Vejle, Denmark
- Department of Oncology, Aarhus University Hospital, Aarhus, Denmark
| | - John Pløen
- Department of Oncology, Vejle Hospital part of Lillebaelt Hospital, Vejle, Denmark
| | - Nina Keldsen
- Department of Oncology, Herning Hospital, Herning, Denmark
| | - Jan Lindebjerg
- Department of Clinical Pathology, Vejle Hospital part of Lillebaelt Hospital, Vejle, Denmark
| | - Flemming Brandt Sørensen
- Institute of Regional Health Research, University of Southern Denmark, Odense, Denmark
- Department of Clinical Pathology, Vejle Hospital part of Lillebaelt Hospital, Vejle, Denmark
| | - Anders Jakobsen
- Department of Oncology, Vejle Hospital part of Lillebaelt Hospital, Vejle, Denmark
- Institute of Regional Health Research, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
36
|
Sebio A, Stintzing S, Stremitzer S, Zhang W, Lenz HJ. Panitumumab: leading to better overall survival in metastatic colorectal cancer? Expert Opin Biol Ther 2014; 14:535-48. [DOI: 10.1517/14712598.2014.894502] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
|
37
|
Boku N, Sugihara K, Kitagawa Y, Hatake K, Gemma A, Yamazaki N, Muro K, Hamaguchi T, Yoshino T, Yana I, Ueno H, Ohtsu A. Panitumumab in Japanese patients with unresectable colorectal cancer: a post-marketing surveillance study of 3085 patients. Jpn J Clin Oncol 2014; 44:214-23. [PMID: 24526771 PMCID: PMC3941645 DOI: 10.1093/jjco/hyt196] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Objective Panitumumab was approved in Japan in April 2010 for the treatment of Kirsten rat sarcoma-2 virus oncogene wild-type unresectable and recurrent colorectal cancer. We conducted a post-marketing surveillance study to evaluate the safety and effectiveness of panitumumab. Methods After panitumumab was commercially available in Japan, all patients to be treated with panitumumab were enrolled. Data on baseline characteristics, treatment outcome, and incidence and severity of adverse drug reactions were collected. Results In total, 3091 patients were registered. In the safety analysis set (n = 3085), panitumumab was administered as monotherapy (40.7%) or combination therapy (59.4%). The median treatment duration was 113 days (range: 1–559 days), and 451 (14.6%) patients received panitumumab for ≥10 months. The overall incidence rate of adverse drug reactions was 84.1%, and the most common adverse drug reaction was skin disorders (78.4%). The incidence rates (all grades) of interstitial lung disease, infusion reaction, electrolyte abnormalities and cardiac disorders were 1.3% (mortality rate: 0.6%), 1.5, 19.3 and 0.2%, respectively. The median survival time of patients treated with panitumumab monotherapy as the third-line, or later, therapy was 10.3 months. Conclusion This post-marketing survey in clinical practice confirmed the safety and effectiveness of panitumumab. The benefit/risk balance for panitumumab in Japanese patients with unresectable colorectal cancer remains favorable.
Collapse
Affiliation(s)
- Narikazu Boku
- *Department of Clinical Oncology, St. Marianna University School of Medicine, 2-16-1, Sugao, Miyamae-ku, Kawasaki 216-8511, Japan.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Aranda F, Vacchelli E, Eggermont A, Galon J, Fridman WH, Zitvogel L, Kroemer G, Galluzzi L. Trial Watch: Immunostimulatory monoclonal antibodies in cancer therapy. Oncoimmunology 2014; 3:e27297. [PMID: 24701370 PMCID: PMC3961485 DOI: 10.4161/onci.27297] [Citation(s) in RCA: 91] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2013] [Accepted: 11/21/2013] [Indexed: 12/16/2022] Open
Abstract
Immunostimulatory monoclonal antibodies (mAbs) exert antineoplastic effects by eliciting a novel or reinstating a pre-existing antitumor immune response. Most often, immunostimulatory mAbs activate T lymphocytes or natural killer (NK) cells by inhibiting immunosuppressive receptors, such as cytotoxic T lymphocyte-associated protein 4 (CTLA4) or programmed cell death 1 (PDCD1, best known as PD-1), or by engaging co-stimulatory receptors, like CD40, tumor necrosis factor receptor superfamily, member 4 (TNFRSF4, best known as OX40) or TNFRSF18 (best known as GITR). The CTLA4-targeting mAb ipilimumab has been approved by the US Food and Drug Administration for use in patients with unresectable or metastatic melanoma in 2011. The therapeutic profile of ipilimumab other CTLA4-blocking mAbs, such as tremelimumab, is currently being assessed in subjects affected by a large panel of solid neoplasms. In the last few years, promising clinical results have also been obtained with nivolumab, a PD-1-targeting mAb formerly known as BMS-936558. Accordingly, the safety and efficacy of nivolumab and other PD-1-blocking molecules are being actively investigated. Finally, various clinical trials are underway to test the therapeutic potential of OX40- and GITR-activating mAbs. Here, we summarize recent findings on the therapeutic profile of immunostimulatory mAbs and discuss clinical trials that have been launched in the last 14 months to assess the therapeutic profile of these immunotherapeutic agents.
Collapse
Affiliation(s)
- Fernando Aranda
- Gustave Roussy; Villejuif, France ; INSERM, U848; Villejuif, France ; Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Centre de Recherche des Cordeliers; Paris, France
| | - Erika Vacchelli
- Gustave Roussy; Villejuif, France ; INSERM, U848; Villejuif, France ; Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Centre de Recherche des Cordeliers; Paris, France ; Université Paris-Sud/Paris XI; Paris, France
| | | | - Jerome Galon
- Université Paris Descartes/Paris V ; Sorbonne Paris Cité; Paris, France ; Université Pierre et Marie Curie/Paris VI; Paris, France ; INSERM, U872; Paris, France ; Equipe 15, Centre de Recherche des Cordeliers; Paris, France
| | - Wolf Hervé Fridman
- Université Pierre et Marie Curie/Paris VI; Paris, France ; INSERM, U872; Paris, France ; Equipe 13, Centre de Recherche des Cordeliers; Paris, France
| | - Laurence Zitvogel
- Gustave Roussy; Villejuif, France ; INSERM, U1015; CICBT507; Villejuif, France
| | - Guido Kroemer
- Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP; Paris, France ; Metabolomics and Cell Biology Platforms; Gustave Roussy; Villejuif, France ; INSERM, U848; Villejuif, France ; Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Centre de Recherche des Cordeliers; Paris, France ; Université Paris Descartes/Paris V ; Sorbonne Paris Cité; Paris, France
| | - Lorenzo Galluzzi
- Gustave Roussy; Villejuif, France ; Université Paris Descartes/Paris V ; Sorbonne Paris Cité; Paris, France ; Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Centre de Recherche des Cordeliers; Paris, France
| |
Collapse
|
39
|
Hocking CM, Townsend AR, Price TJ. Panitumumab in metastatic colorectal cancer. Expert Rev Anticancer Ther 2014; 13:781-93. [DOI: 10.1586/14737140.2013.811064] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
40
|
|
41
|
A Phase 2 Randomized Trial of Paclitaxel and Carboplatin with or without Panitumumab for First-Line Treatment of Advanced Non–Small-Cell Lung Cancer. J Thorac Oncol 2013; 8:1510-8. [DOI: 10.1097/jto.0b013e3182a7d1da] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
|
42
|
Pharmacokinetics of panitumumab in a patient with liver dysfunction: a case report. Cancer Chemother Pharmacol 2013; 73:429-33. [PMID: 24258455 DOI: 10.1007/s00280-013-2353-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2013] [Accepted: 11/07/2013] [Indexed: 12/18/2022]
Abstract
PURPOSE Panitumumab is used for the treatment for metastatic RAS wild-type colorectal cancer (mCRC). It is likely that many of these patients will present with liver metastases and some with liver dysfunction. The pharmacokinetics in patients with hepatic impairment has not been investigated, and dosage adjustments are undetermined. Here, we present a case of a patient with progressive mCRC and liver dysfunction. METHODS A heavily pretreated KRAS wild-type mCRC patient with liver disease Child-Pugh class B was treated with 2-weekly intravenous panitumumab (6 mg/kg). The patient received 2 doses of 490 mg i.v. panitumumab after which progressive disease was documented. Toxicities were graded using CTCAEv4.0. Serum samples were collected, and panitumumab concentrations were determined using a validated immunoassay. Pharmacokinetic parameters after the first dose, including dose-normalized AUC from time zero-day 14, clearance (CL), and elimination half-life (T1/2), were estimated via trapezoidal noncompartmental methods. Data were compared to historical data from a population with adequate liver function, as reported by Stephenson (Clin Colorectal Cancer, 8:29-37, 2009). Values within the range of the mean ±1 standard deviation (SD) were considered not deviant. RESULTS Calculated AUC after the first dose of 6 mg/kg panitumumab in this patient with hepatic dysfunction was 877 μg day/mL (Stephenson's cohort 1: 744 ± 195 μg day/mL). Estimated T1/2 was 3.58 days (5.28 ± 1.90 days), and CL was 6.9 mL/day/kg (8.21 ± 3.79 mL/day/kg). Estimated PK parameters during the first cycle were inside reported mean ±1 SD of historical controls without liver dysfunction. No toxicity was reported during treatment; particularly, no diarrhea and skin toxicity were noticed. CONCLUSIONS The pharmacokinetics of panitumumab in this patient suffering from metastatic colorectal cancer with liver dysfunction Child-Pugh class B was similar compared to patients with adequate liver function. Moreover, no substantial toxicity was detected. The here-presented data may help clinical decision making in real-life practice. Two-weekly panitumumab monotherapy seems to be safely applicable in patients with KRAS wild-type mCRC and hepatic dysfunction, without the need for any dose adjustments.
Collapse
|
43
|
Gao M, Liang XJ, Zhang ZS, Ma W, Chang ZW, Zhang MZ. Relationship between expression of EGFR in gastric cancer tissue and clinicopathological features. ASIAN PAC J TROP MED 2013; 6:260-4. [PMID: 23608326 DOI: 10.1016/s1995-7645(13)60054-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2013] [Revised: 02/15/2012] [Accepted: 03/15/2012] [Indexed: 11/30/2022] Open
Abstract
OBJECTIVE To investigate the relationship between the expression of epidermal growth factor receptor (EGFR) in gastric cancer and the clinicopathological features and prognosis. METHODS A total of 78 paraffin specimens of gastric cancer operation were collected. The immunohistochemical method was used to detect the expression of EGFR in 78 cases of gastric cancer and 20 cases of adjacent normal tissue. The relationship between the high expression of EGFR and clinicopathological features was analyzed. RESULTS EGFR positive expression rate in the 78 cases of gastric cancer tissue was 57.7 % (45/78), while EGFR was not expressed in 20 cases of adjacent normal tissue. The high EGFR expression was positively correlated with the position of gastric cancer, tumor size, cell differentiation, invasive depth, lymph node metastasis and TNM staging, yet having no obvious relation with gender or age. CONCLUSIONS EGFR expression level in gastric cancer is closely related to the incidence and development of gastric cancer, which can provide a theoretical basis for the targeted therapy for gastric cancer with EGFR as the target.
Collapse
Affiliation(s)
- Ming Gao
- Oncology Department, First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China.
| | | | | | | | | | | |
Collapse
|
44
|
Zhang J, Hochwald SN. Targeting Receptor Tyrosine Kinases in Solid Tumors. Surg Oncol Clin N Am 2013; 22:685-703. [DOI: 10.1016/j.soc.2013.06.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
45
|
Cananzi FCM, Jayanth A, Lorenzi B, Belgaumkar A, Mochlinski K, Sharma A, Mudan S, Cunningham D. "Chronic" metastatic pancreatic acinar cell carcinoma. Pancreatology 2013; 13:549-52. [PMID: 24075523 DOI: 10.1016/j.pan.2013.05.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2013] [Revised: 04/13/2013] [Accepted: 05/02/2013] [Indexed: 12/11/2022]
Abstract
Acinar cell carcinoma (ACC) of the pancreas is a rare exocrine tumour for which there is very limited information about chemotherapy regimens and prognosis. Even though there are clinical guidelines for management of ductal cell carcinoma, a definitive and specific regime has not yet been agreed for this type of pancreatic cancer. We report a case of metastatic ACC of pancreas who has been treated with a multimodal approach, including novel combinations of different targeted drugs with conventional chemotherapy, surgery and radiofrequency ablation since the last 11 years. This degree of long term survival has not been reported so far in such a case of metastatic ACC of the pancreas. This case highlights the importance of a personalised multidisciplinary therapeutic strategy, employing locoregional therapies along with combinations of established and novel systemic therapies to control the disease, and the importance of flexibility when instigating new treatment paradigms for progressive cancer. Also, this case demonstrates that complete tumour eradication may not be the sole purpose of surgical oncology.
Collapse
|
46
|
Ocana A, Amir E, Vera-Badillo F, Seruga B, Tannock IF. Phase III trials of targeted anticancer therapies: redesigning the concept. Clin Cancer Res 2013; 19:4931-40. [PMID: 23881926 DOI: 10.1158/1078-0432.ccr-13-1222] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Randomized phase III trials provide the gold-standard evidence for the approval of new drugs: an experimental treatment is compared with the current standard of care to identify clinically relevant differences in a predefined endpoint. However, there are several problems relating to the current role of phase III trials in drug development including the limited clinical benefit observed for some approved agents, the necessity for large trials to detect these differences, the inability of such trials to identify rare but important toxicities, and high cost. The design of phase III trials evaluating drug combinations, and those including biomarkers, presents additional challenges. Here, we review these problems and suggest that phase III trials with adaptive designs in selected prescreened populations could reduce these limitations.
Collapse
Affiliation(s)
- Alberto Ocana
- Authors' Affiliation: Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre and the University of Toronto, Toronto, Canada
| | | | | | | | | |
Collapse
|
47
|
Abstract
Pancreatic cancer is the fourth leading cause of cancer-related death. Most patients present with an advanced stage of disease that has a dismal outcome, with a median survival of approximately 6 months. Evidently, there is a clear need for the development of new agents with novel mechanisms of action in this disease. A number of biological agents modulating different signal transduction pathways are currently in clinical development, inhibiting angiogenesis and targeting epidermal growth factor receptor, cell cycle, matrix metalloproteinases, cyclooxygenase-2, mammalian target of rapamycin, or proteasome. This is the first systematic review of the literature to synthesize all available data coming from trials and evaluate the efficacy and safety of molecular targeted drugs in unresectable and metastatic pancreatic cancer. However, it should be stressed that although multiple agents have been tested, only 9 phase 3 trials have been conducted and one agent (erlotinib) has been approved by the Food and Drug Administration for use in clinical practice. As knowledge accumulates on the molecular mechanisms underlying carcinogenesis in the pancreas, the anticipated development and assessment of molecularly targeted agents may offer a promising perspective for a disease which, to date, remains incurable.
Collapse
|
48
|
Seymour MT, Brown SR, Middleton G, Maughan T, Richman S, Gwyther S, Lowe C, Seligmann JF, Wadsley J, Maisey N, Chau I, Hill M, Dawson L, Falk S, O'Callaghan A, Benstead K, Chambers P, Oliver A, Marshall H, Napp V, Quirke P. Panitumumab and irinotecan versus irinotecan alone for patients with KRAS wild-type, fluorouracil-resistant advanced colorectal cancer (PICCOLO): a prospectively stratified randomised trial. Lancet Oncol 2013; 14:749-59. [PMID: 23725851 PMCID: PMC3699713 DOI: 10.1016/s1470-2045(13)70163-3] [Citation(s) in RCA: 296] [Impact Index Per Article: 26.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Background Therapeutic antibodies targeting EGFR have activity in advanced colorectal cancer, but results from clinical trials are inconsistent and the population in which most benefit is derived is uncertain. Our aim was to assess the addition of panitumumab to irinotecan in pretreated advanced colorectal cancer. Methods In this open-label, randomised trial, we enrolled patients who had advanced colorectal cancer progressing after fluoropyrimidine treatment with or without oxaliplatin from 60 centres in the UK. From December, 2006 until June, 2008, molecularly unselected patients were recruited to a three-arm design including irinotecan (control), irinotecan plus ciclosporin, and irinotecan plus panitumumab (IrPan) groups. From June 10, 2008, in response to new data, the trial was amended to a prospectively stratified design, restricting panitumumab randomisation to patients with KRAS wild-type tumours; the results of the comparison between the irinotcan and IrPan groups are reported here. We used a computer-generated randomisation sequence (stratified by previous EGFR targeted therapy and then minimised by centre, WHO performance status, previous oxaliplatin, previous bevacizumab, previous dose modifications, and best previous response) to randomly allocate patients to either irinotecan or IrPan. Patients in both groups received 350 mg/m2 intravenous irinotecan every 3 weeks (300 mg/m2 if aged ≥70 years or a performance status of 2); patients in the IrPan group also received intravenous panitumumab 9 mg/kg every 3 weeks. The primary endpoint was overall survival in KRAS wild-type patients who had not received previous EGFR targeted therapy, analysed by intention to treat. Tumour DNA was pyrosequenced for KRASc.146, BRAF, NRAS, and PIK3CA mutations, and predefined molecular subgroups were analysed for interaction with the effect of panitumumab. This study is registered, number ISRCTN93248876. Results Between Dec 4, 2006, and Aug 31, 2010, 1198 patients were enrolled, of whom 460 were included in the primary population of patients with KRASc.12–13,61 wild-type tumours and no previous EGFR targeted therapy. 230 patients were randomly allocated to irinotecan and 230 to IrPan. There was no difference in overall survival between groups (HR 1·01, 95% CI 0·83–1·23; p=0·91), but individuals in the IrPan group had longer progression-free survival (0·78, 0·64–0·95; p=0·015) and a greater number of responses (79 [34%] patients vs 27 [12%]; p<0·0001) than did individuals in the irinotecan group. Grade 3 or worse diarrhoea (64 [29%] of 219 patients vs 39 [18%] of 218 patients), skin toxicity (41 [19%] vs none), lethargy (45 [21]% vs 24 [11%]), infection (42 [19%] vs 22 [10%]) and haematological toxicity (48 [22%] vs 27 [12%]) were reported more commonly in the IrPan group than in the irinotecan group. We recorded five treatment-related deaths, two in the IrPan group and three in the irinotecan group. Interpretation Adding panitumumab to irinotecan did not improve the overall survival of patients with wild-type KRAS tumours. Further refinement of molecular selection is needed for substantial benefits to be derived from EGFR targeting agents. Funding Cancer Research UK, Amgen Inc.
Collapse
|
49
|
Panitumumab and irinotecan every 3 weeks is an active and convenient regimen for second-line treatment of patients with wild-type K-RAS metastatic colorectal cancer. Clin Transl Oncol 2013; 15:705-11. [DOI: 10.1007/s12094-012-0993-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2012] [Accepted: 12/20/2012] [Indexed: 12/27/2022]
|
50
|
Cathomas R, Rothermundt C, Klingbiel D, Bubendorf L, Jaggi R, Betticher DC, Brauchli P, Cotting D, Droege C, Winterhalder R, Siciliano D, Berthold DR, Pless M, Schiess R, von Moos R, Gillessen S. Efficacy of cetuximab in metastatic castration-resistant prostate cancer might depend on EGFR and PTEN expression: results from a phase II trial (SAKK 08/07). Clin Cancer Res 2012; 18:6049-57. [PMID: 22977195 DOI: 10.1158/1078-0432.ccr-12-2219] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
PURPOSE The EGF receptor (EGFR) is overexpressed in the majority of metastatic castration-resistant prostate cancers (mCRPC) and might represent a valid therapeutic target. The combination of docetaxel and cetuximab, the monoclonal antibody against EGFR, has not been tested in patients with prostate cancer. EXPERIMENTAL DESIGN Patients with mCRPC progressing during or within 90 days after at least 12 weeks of docetaxel were included in this phase II trial. Treatment consisted of docetaxel (75 mg/m(2) every 3 weeks or 35 mg/m(2) on days 1, 8, 15 every 4 weeks) in combination with cetuximab (400 mg/m(2) on day 1 and then 250 mg/m(2) weekly). The primary endpoint was progression-free survival (PFS) at 12 weeks defined as the absence of prostate-specific antigen (PSA), radiographic, or clinical progression. Evaluation of known biomarkers of response and resistance to cetuximab (EGFR, PTEN, amphiregulin, epiregulin) was conducted. RESULTS Thirty-eight patients were enrolled at 15 Swiss centers. Median age was 68 years and median PSA was 212 ng/mL. PFS at 12 weeks was 34% [95% confidence interval (CI), 19%-52%], PFS at 24 weeks was 20%, and median overall survival (OS) was 13.3 months (95% CI, 7.3-15.4). Seven patients (20%) had a confirmed ≥ 50% and 11 patients (31%) a confirmed ≥ 30% PSA decline. About 47% of enrolled patients experienced grade 3 and 8% grade 4 toxicities. A significantly improved PFS was found in patients with overexpression of EGFR and persistent activity of PTEN. CONCLUSIONS EGFR inhibition with cetuximab might improve the outcome of patients with mCRPC. A potential correlation between EGFR overexpression, persistent expression of PTEN, and EGFR inhibition should be investigated further.
Collapse
|