1
|
Davidson TM, Lebreton CL, Hendricksen AEW, Atkinson HJ, Larson MC, Oberg AL, Provencher DM, Glaspy JA, Karlan BY, Slamon DJ, Konecny GE, Ray-Coquard IL. Results of TRIO-15, a multicenter, open-label, phase II study of the efficacy and safety of ganitumab in patients with recurrent platinum-sensitive ovarian cancer. Gynecol Oncol 2023; 170:221-228. [PMID: 36709663 PMCID: PMC10425916 DOI: 10.1016/j.ygyno.2023.01.021] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Revised: 01/14/2023] [Accepted: 01/17/2023] [Indexed: 01/30/2023]
Abstract
BACKGROUND IGF signaling has been implicated in the pathogenesis and progression of ovarian carcinoma (OC). Single agent activity and safety of ganitumab (AMG 479), a fully human monoclonal antibody against IGF1R that blocks binding of IGF1 and IGF2, were evaluated in patients with platinum-sensitive recurrent OC. METHODS Patients with CA125 progression (GCIG criteria) or measurable disease per RECIST following primary platinum-based therapy received 18 mg/kg of ganitumab q3w. The primary endpoint was objective response rate (ORR) assessed per RECIST 1.1 by an independent radiology review committee (IRC) and/or GCIG CA125 criteria. Secondary endpoints included clinical benefit rate (CBR), progression free survival (PFS) and overall survival (OS). RESULTS 61 pts. were accrued. Objective responses were seen in 5/61 patients (ORR 8.2%, 95% CI, 3.1-18.8) with 1 partial response (PR) by RECIST and 2 complete responses (CR) as well as 2 PR by CA125 criteria. CBR was 80.3% (95% CI, 67.8-89.0%). The median PFS according to RECIST by IRC was 2.1 months (95% CI, 2.0-3.1). The median PFS per RECIST IRC and/or CA125 was 2.0 months (95% CI, 1.8-2.2). The median OS was 21 months (95% CI, 19.5-NA). The most common overall adverse events were fatigue (36.1%) and hypertension (34.4%). Grade 1/2 hyperglycemia occurred in 30.4% of patients. Hypertension (11.5%) and hypersensitivity (8.2%) were the most frequent grade 3 adverse events. CONCLUSIONS IGF1R inhibition with ganitumab was well-tolerated, however, our results do not support further study of ganitumab as a single agent in unselected OC patients.
Collapse
Affiliation(s)
- T M Davidson
- Division of Oncology, Mayo Clinic, Rochester, MN, USA
| | | | | | - H J Atkinson
- Division of Clinical Trials and Biostatistics, Department of Quantitative Health Science, Mayo Clinic, Rochester, MN, USA
| | - M C Larson
- Division of Clinical Trials and Biostatistics, Department of Quantitative Health Science, Mayo Clinic, Rochester, MN, USA
| | - A L Oberg
- Division of Computational Biology, Department of Quantitative Health Science, Mayo Clinic, Rochester, MN, USA
| | | | - J A Glaspy
- Division of Hematology/Oncology, University of California Los Angeles, Los Angeles, CA, USA
| | - B Y Karlan
- Division of Gynecologic Oncology, University of California Los Angeles, Los Angeles, CA, USA
| | - D J Slamon
- Division of Hematology/Oncology, University of California Los Angeles, Los Angeles, CA, USA
| | - G E Konecny
- Division of Hematology/Oncology, University of California Los Angeles, Los Angeles, CA, USA; Division of Gynecologic Oncology, University of California Los Angeles, Los Angeles, CA, USA.
| | - I L Ray-Coquard
- Centre Léon Bérard, Lyon, France; Health Services and Performance Research Lab (EA 7425 HESPER), University Claude Bernard Lyon 1, 69008 Lyon, France
| |
Collapse
|
2
|
Liu Y, Yu S, Xu T, Bodenko V, Orlova A, Oroujeni M, Rinne SS, Tolmachev V, Vorobyeva A, Gräslund T. Preclinical Evaluation of a New Format of 68Ga- and 111In-Labeled Affibody Molecule Z IGF-1R:4551 for the Visualization of IGF-1R Expression in Malignant Tumors Using PET and SPECT. Pharmaceutics 2022; 14:pharmaceutics14071475. [PMID: 35890370 PMCID: PMC9320461 DOI: 10.3390/pharmaceutics14071475] [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: 06/22/2022] [Revised: 07/11/2022] [Accepted: 07/13/2022] [Indexed: 02/04/2023] Open
Abstract
The Insulin-like growth factor-1 receptor (IGF-1R) is a molecular target for several monoclonal antibodies undergoing clinical evaluation as anticancer therapeutics. The non-invasive detection of IGF-1R expression in tumors might enable stratification of patients for specific treatment and improve the outcome of both clinical trials and routine treatment. The affibody molecule ZIGF-1R:4551 binds specifically to IGF-1R with subnanomolar affinity. The goal of this study was to evaluate the 68Ga and 111In-labeled affibody construct NODAGA-(HE)3-ZIGF-1R:4551 for the imaging of IGF-1R expression, using PET and SPECT. The labeling was efficient and provided stable coupling of both radionuclides. The two imaging probes, [68Ga]Ga-NODAGA-(HE)3-ZIGF-1R:4551 and [111In]In-NODAGA-(HE)3-ZIGF-1R:4551, demonstrated specific binding to IGF-1R-expressing human cancer cell lines in vitro and to IGF-1R-expressing xenografts in mice. Preclinical PET and SPECT/CT imaging demonstrated visualization of IGF-1R-expressing xenografts already one hour after injection. The tumor-to-blood ratios at 3 h after injection were 7.8 ± 0.2 and 8.0 ± 0.6 for [68Ga]Ga-NODAGA-(HE)3-ZIGF-1R:4551 and [111In]In-NODAGA-(HE)3-ZIGF-1R:4551, respectively. In conclusion, a molecular design of the ZIGF-1R:4551 affibody molecule, including placement of a (HE)3-tag on the N-terminus and site-specific coupling of a NODAGA chelator on the C-terminus, provides a tracer with improved imaging properties for visualization of IGF-1R in malignant tumors, using PET and SPECT.
Collapse
Affiliation(s)
- Yongsheng Liu
- Department of Immunology, Genetics and Pathology, Uppsala University, 75237 Uppsala, Sweden; (Y.L.); (T.X.); (M.O.); (A.V.)
| | - Shengze Yu
- Department of Protein Science, KTH Royal Institute of Technology, 10044 Stockholm, Sweden;
| | - Tianqi Xu
- Department of Immunology, Genetics and Pathology, Uppsala University, 75237 Uppsala, Sweden; (Y.L.); (T.X.); (M.O.); (A.V.)
| | - Vitalina Bodenko
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, 634050 Tomsk, Russia; (V.B.); (A.O.)
| | - Anna Orlova
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, 634050 Tomsk, Russia; (V.B.); (A.O.)
- Department of Medicinal Chemistry, Uppsala University, 75123 Uppsala, Sweden;
| | - Maryam Oroujeni
- Department of Immunology, Genetics and Pathology, Uppsala University, 75237 Uppsala, Sweden; (Y.L.); (T.X.); (M.O.); (A.V.)
- Affibody AB, 17165 Solna, Sweden
| | - Sara S. Rinne
- Department of Medicinal Chemistry, Uppsala University, 75123 Uppsala, Sweden;
| | - Vladimir Tolmachev
- Department of Immunology, Genetics and Pathology, Uppsala University, 75237 Uppsala, Sweden; (Y.L.); (T.X.); (M.O.); (A.V.)
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, 634050 Tomsk, Russia; (V.B.); (A.O.)
- Correspondence: (V.T.); (T.G.); Tel.: +46-704-250782 (V.T.); +46-8790-9627 (T.G.)
| | - Anzhelika Vorobyeva
- Department of Immunology, Genetics and Pathology, Uppsala University, 75237 Uppsala, Sweden; (Y.L.); (T.X.); (M.O.); (A.V.)
| | - Torbjörn Gräslund
- Department of Protein Science, KTH Royal Institute of Technology, 10044 Stockholm, Sweden;
- Correspondence: (V.T.); (T.G.); Tel.: +46-704-250782 (V.T.); +46-8790-9627 (T.G.)
| |
Collapse
|
3
|
Konecny GE, Hendrickson AEW, Davidson TM, Winterhoff BJ, Ma S, Mahner S, Sehouli J, Fasching PA, Feisel-Schwickardi G, Poelcher M, Roman LD, Rody A, Karlan BY, Mullany SA, Chen H, Ray-Coquard IL, Provencher DM, Yachnin A, Cottu PH, Glaspy JA, Haluska P, Slamon DJ. Results of TRIO-14, a phase II, multicenter, randomized, placebo-controlled trial of carboplatin-paclitaxel versus carboplatin-paclitaxel-ganitumab in newly diagnosed epithelial ovarian cancer. Gynecol Oncol 2021; 163:465-472. [PMID: 34642026 DOI: 10.1016/j.ygyno.2021.09.025] [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: 07/16/2021] [Revised: 09/03/2021] [Accepted: 09/07/2021] [Indexed: 12/28/2022]
Abstract
PURPOSE Insulin-like growth factor (IGF) signaling is implicated in pathogenesis and chemotherapy resistance of epithelial ovarian cancer (EOC). We explored efficacy and safety of adding ganitumab, a monoclonal antibody targeting IGF-1R, to carboplatin/paclitaxel (CP) chemotherapy in patients with primary EOC. DESIGN Patients were randomly assigned to receive CP/ganitumab (18 mg/kg q3w) or CP/placebo for 6 cycles followed by 6 cycles of single agent ganitumab/placebo maintenance therapy as front-line therapy. Primary endpoint was progression free survival. Secondary endpoints were time to progression and overall survival. Pretreatment samples were prospectively collected for retrospective biomarker analyses. RESULTS 170 patients enrolled. 165 patients assessable for toxicity. Median PFS was 15.7 months with CP/ganitumab and 16.7 months with CP/placebo (HR 1.23; 95% CI 0.82-1.83, P = 0.313). All grade neutropenia (84.1% vs 71.4%), thrombocytopenia (75.3% vs 57.1%) and hyperglycemia (15.9% vs 2.6%) were more common in the ganitumab group compared to the placebo group. Ganitumab/placebo related serious adverse events were reported in 26.1% of the patients with ganitumab and in 6.5% with placebo. Non-progression related fatal events were more common with ganitumab (5 versus 2 patients). The ganitumab group experienced more dose delays which resulted in lower relative dose intensity of chemotherapy in the experimental group. In an exploratory model IGFBP2 expression was predictive of ganitumab response (treatment interaction; PFS, P = 0.03; OS, P = 0.01). CONCLUSION Addition of ganitumab to CP chemotherapy in primary EOC did not improve PFS. Our results do not support further study of ganitumab in unselected EOC patients.
Collapse
Affiliation(s)
- G E Konecny
- Division of Hematology/Oncology, University of California Los Angeles, Los Angeles, CA, USA; Division of Gynecologic Oncology, University of California Los Angeles, Los Angeles, CA, USA.
| | | | - T M Davidson
- Division of Oncology Mayo Clinic, Rochester, MN, USA
| | - B J Winterhoff
- Department of Obstetrics and Gynecology, University of Minnesota, Minneapolis, MN, USA
| | - S Ma
- Institute for Health Informatics, School of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - S Mahner
- Department of Gynecology and Gynecologic Oncology, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - J Sehouli
- University Hospital Charite, Campus Virchow-Klinikum, Klinik für Frauenheilkunde und Geburtshilfe & Nord-Ostdeutsche-Gesellschaft für Gynäkologische Onkologie (NOGGO), Berlin, Germany
| | - P A Fasching
- Department of Obstetrics and Gynecology, University of Erlangen, Erlangen, Germany
| | | | - M Poelcher
- Department of Gynecology, Rotkreutzklinikum, Munich, Germany
| | - L D Roman
- USC Norris Comprehensive Cancer Center, Los Angeles, CA, USA
| | - A Rody
- Department of Obstetrics and Gynecology, Universitätsklinikum Schleswig-Holstein, Campus Lübeck, Germany
| | - B Y Karlan
- Division of Gynecologic Oncology, University of California Los Angeles, Los Angeles, CA, USA
| | - S A Mullany
- Department of Obstetrics and Gynecology, University of Minnesota, Minneapolis, MN, USA
| | - H Chen
- Division of Hematology/Oncology, University of California Los Angeles, Los Angeles, CA, USA
| | | | | | - A Yachnin
- Department of Oncology, Kaplan Medical Center, Rehovot, Israel
| | | | - J A Glaspy
- Division of Hematology/Oncology, University of California Los Angeles, Los Angeles, CA, USA
| | - P Haluska
- Bristol-Myers Squibb Inc, Lawrenceville, NJ, USA
| | - D J Slamon
- Division of Hematology/Oncology, University of California Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
4
|
Amelot A, Terrier LM, Cristini J, Buffenoir K, Pascal-Moussellard H, Carpentier A, Bonaccorsi R, Le Nail LR, Mathon B. Spinal metastases from lung cancer: Survival depends only on genotype, neurological and personal status, scarcely of surgical resection. Surg Oncol 2020; 34:51-56. [PMID: 32891353 DOI: 10.1016/j.suronc.2020.03.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 01/30/2020] [Accepted: 03/30/2020] [Indexed: 12/11/2022]
Abstract
BACKGROUND For patients with non-small cell lung cancer (NSCLC), the spinal column is the most common site for bone metastasis. Studies that assess survival prognostic factors associated with specific lung spinal metastases (SpM) are weak and required the incorporation of genotype mutations. METHODS A prospective French national multicenter database of patients treated for SpM between January 2014 and 2017.818 lung SpM were diagnosed over the course or at the time of diagnosis of 210 consecutive patients with NSCLC. RESULTS Median overall survival (OS) time for all patients from the lung SpM event was 5.9 months (SD 0.609). For 122 patients (61%), lung tumor and SpM were diagnosed synchronously. In univariate analysis, good World Health Organisation (WHO) status (p < 0.0001), ambulatory status (Frankel score) (p < 0.0001), the absence of spine epiduritis (p < 0.0001), immunotherapy after SpM diagnosis (p < 0.0001), ALK gene rearrangement (p < 0.0001) and EGFR mutation (p < 0.0001) were associated with longer survival, whereas spine surgery showed no association (0.141). Cox multivariate proportional hazard model identified that EGFR + status (HR: 0.339, 95% CI 0.166-0.693; p = 0.003), good WHO status (p < 0.0001) and good neurological status (Frankel E; p < 0.001 and D; p = 0.018) were associated with higher median OS. Whereas the other factors, including ALK + status, epiduritis and immunotherapy were not independent prognostic factors of survival. CONCLUSION Survival in SpM must be prognosticated from general health performance status: clinical (WHO) and neurological (Frankel) as well as the EGFR mutation status. Immunotherapy, surgery and epiduritis have not demonstrated prognostic value. Therefore, surgical prognostic scoring algorithms should incorporate genotype subtypes in NSCLC cancers to adapt surgical treatment.
Collapse
Affiliation(s)
- Aymeric Amelot
- Department of Neurosurgery, La Pitié Salpétrière Hospital, Paris, France.
| | | | - Joseph Cristini
- Department of Neurosurgery/Neurotraumatology, Hotel-Dieu Hospital, Nantes, France
| | - Kévin Buffenoir
- Department of Neurosurgery/Neurotraumatology, Hotel-Dieu Hospital, Nantes, France
| | | | | | - Raphael Bonaccorsi
- Department of Orthopaedic Surgery, Pitié-Salpêtrière Hospital, Paris, France
| | | | - Bertrand Mathon
- Department of Neurosurgery, La Pitié Salpétrière Hospital, Paris, France
| |
Collapse
|
5
|
Ye Z, Zhang B, Chen Y, Lin J. Comparison of single utility port video-assisted thoracoscopic surgery (VATS) and three-port VATS for non-small cell lung cancer. Oncol Lett 2019; 18:1311-1317. [PMID: 31423191 PMCID: PMC6607390 DOI: 10.3892/ol.2019.10394] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Accepted: 04/24/2019] [Indexed: 11/11/2022] Open
Abstract
Clinical efficacy of single utility port video-assisted thoracoscopic surgery (VATS) and three-port VATS for patients with non-small cell lung cancer (NSCLC) was compared. A total of 156 patients with NSCLC who underwent VATS in Taizhou Hospital of Zhejiang Province from July 2015 to January 2017 were selected as subjects. They were randomly divided into group A (n=74) and group B (n=82), in which group A was treated with single utility port VATS and group B was treated with three-port VATS. Perioperative indicators such as operation time, intraoperative blood loss, postoperative drainage, removal of drainage tube, lymph node dissection, hospitalization time, postoperative complications, postoperative pain and postoperative quality of life were observed. Expression levels of CRP and IL-6 in serum were detected by enzyme-linked immunosorbent assay (ELISA). There was no significant difference in the operation time, postoperative drainage volume, drainage tube removal time and lymph node dissection between groups A and B (P>0.05). Blood loss and hospitalization time in group A were significantly lower than those in group B (P<0.001). VAS scores at 1–3 days after operation in group A were significantly lower than those in group B (P<0.001). Levels of serum CRP and IL-6 at 1–7 days after operation in group A were significantly lower than those in group B (P<0.001). Incidence of complication in group A was not significantly different from that in group B (P>0.05). Overall quality of life scores of group A and B were significantly lower than the preoperative scores (P<0.001). Overall status score of group A was significantly higher than that of group B (P<0.001). Clinical efficacies of single utility port VATS and three-port VATS were similar. Single utility port VATS can reduce trauma during surgery, reduce stress response, relieve postoperative pain, and facilitate the recovery of postoperative quality of life.
Collapse
Affiliation(s)
- Zhongrui Ye
- Department of Thoracic Surgery, Taizhou Hospital of Zhejiang Province, Taizhou, Zhejiang 317000, P.R. China
| | - Bo Zhang
- Department of Thoracic Surgery, Taizhou Hospital of Zhejiang Province, Taizhou, Zhejiang 317000, P.R. China
| | - Yu Chen
- Department of Thoracic Surgery, Taizhou Hospital of Zhejiang Province, Taizhou, Zhejiang 317000, P.R. China
| | - Jiang Lin
- Department of Thoracic Surgery, Taizhou Hospital of Zhejiang Province, Taizhou, Zhejiang 317000, P.R. China
| |
Collapse
|
6
|
Rieunier G, Wu X, Macaulay VM, Lee AV, Weyer-Czernilofsky U, Bogenrieder T. Bad to the Bone: The Role of the Insulin-Like Growth Factor Axis in Osseous Metastasis. Clin Cancer Res 2019; 25:3479-3485. [PMID: 30745299 DOI: 10.1158/1078-0432.ccr-18-2697] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 01/10/2019] [Accepted: 02/06/2019] [Indexed: 11/16/2022]
Abstract
Bone metastases are a frequent complication of cancer that are associated with considerable morbidity. Current treatments may temporarily palliate the symptoms of bone metastases but often fail to delay their progression. Bones provide a permissive environment because they are characterized by dynamic turnover, secreting factors required for bone maintenance but also stimulating the establishment and growth of metastases. Insulin-like growth factors (IGF) are the most abundant growth factors in bone and are required for normal skeletal development and function. Via activation of the IGF-1 receptors (IGF-1R) and variant insulin receptors, IGFs promote cancer progression, aggressiveness, and treatment resistance. Of specific relevance to bone biology, IGFs contribute to the homing, dormancy, colonization, and expansion of bone metastases. Furthermore, preclinical evidence suggests that tumor cells can be primed to metastasize to bone by a high IGF-1 environment in the primary tumor, suggesting that bone metastases may reflect IGF dependency. Therapeutic targeting of the IGF axis may therefore provide an effective method for treating bone metastases. Indeed, anti-IGF-1R antibodies, IGF-1R tyrosine kinase inhibitors, and anti-IGF-1/2 antibodies have demonstrated antitumor activity in preclinical models of prostate and breast cancer metastases, either alone or in combination with other agents. Several studies suggest that such treatments can inhibit bone metastases without affecting growth of the primary tumor. Although previous trials of anti-IGF-1R drugs have generated negative results in unselected patients, these considerations suggest that future clinical trials of IGF-targeted agents may be warranted in patients with bone metastases.
Collapse
Affiliation(s)
| | - Xiaoning Wu
- Department of Oncology, University of Oxford, Oxford, United Kingdom
| | | | - Adrian V Lee
- Department of Pharmacology and Chemical Biology, UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, Pennsylvania
| | | | - Thomas Bogenrieder
- RCV Medicine, Boehringer Ingelheim RCV, Vienna, Austria.,Department of Urology, University Hospital Grosshadern, Ludwig-Maximilians-University, Munich, Germany
| |
Collapse
|
7
|
Abstract
INTRODUCTION Epidermal Growth Factor Receptor (EGFR)-dependent signaling plays a crucial role in epithelial cancer biology, and dictated the development of several targeting agents. The mouse-human chimeric antibody Cetuximab was among the first to be developed. After about two decades of clinical research it has gained a significant place in the management of advanced colorectal and head and neck cancers, whereas its development in non small cell lung cancer (NSCLC) has not led to a place in routine clinical practice, because of marginal clinical benefit despite statistically significant Phase III trials. Recent data from ongoing trials suggest that more careful selection based on molecular markers may identify good responders. Areas covered: In this article, the authors review the literature concerning basic science studies identifying EGFR as a therapeutic target, pharmacological development of Cetuximab, its pharmacodynamics and pharmacokinetics, and clinical trials on Cetuximab in NSCLC, focusing on recent findings on putative predictive biomarkers. Expert opinion: Cetuximab currently has no role in NSCLC treatment outside of research settings. We argue that failure to identify a predictive biomarker early on has hampered its chances to enter routine practice. Although recent research suggests benefit in highly selected patient subsets, its potential impact is severely dampened by lack of regulatory body approval and the emergence of competitors for the same niches.
Collapse
Affiliation(s)
- Luca Mazzarella
- a University of Milano, Department of Oncology and Hemato-Oncology, Division of Early Drug Development for Innovative Therapies , European Institute of Oncology , Milano , Italia
| | - Alessandro Guida
- a University of Milano, Department of Oncology and Hemato-Oncology, Division of Early Drug Development for Innovative Therapies , European Institute of Oncology , Milano , Italia
| | - Giuseppe Curigliano
- a University of Milano, Department of Oncology and Hemato-Oncology, Division of Early Drug Development for Innovative Therapies , European Institute of Oncology , Milano , Italia
| |
Collapse
|
8
|
Agustoni F, Suda K, Yu H, Ren S, Rivard CJ, Ellison K, Caldwell C, Rozeboom L, Brovsky K, Hirsch FR. EGFR-directed monoclonal antibodies in combination with chemotherapy for treatment of non-small-cell lung cancer: an updated review of clinical trials and new perspectives in biomarkers analysis. Cancer Treat Rev 2018; 72:15-27. [PMID: 30445271 DOI: 10.1016/j.ctrv.2018.08.002] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Revised: 07/30/2018] [Accepted: 08/03/2018] [Indexed: 02/08/2023]
Abstract
Lung cancer still represents one of the most common and fatal neoplasm, accounting for nearly 30% of all cancer-related deaths. Targeted therapies based on molecular tumor features and programmed death-1 (PD-1)/programmed death ligand-1 (PDL-1) blockade immunotherapy have offered new therapeutic options for patients with advanced non-small-cell lung cancer (NSCLC). Activation of the epidermal growth factor receptor (EGFR)-pathway promotes tumor growth and progression, including angiogenesis, invasion, metastasis and inhibition of apoptosis, providing a strong rationale for targeting this pathway. EGFR expression is detected in up to 85% of NSCLC and has been demonstrated to be associated with poor prognosis. Two approaches for blocking EGFR signaling are available: prevention of ligand binding to the extracellular domain with monoclonal antibodies (mAbs) and inhibition of the intracellular tyrosine kinase activity with small molecules. There is a strong rationale to consider the tumor's level of EGFR expression as one of the most significant predictive biomarkers in this setting. In this paper we provide an update focusing on the current status of EGFR-directed mAbs use for the treatment of patients with advanced NSCLC, through a review of all clinical trials involving anti-EGFR mAbs in combination with chemotherapy (CT) for advanced disease and with chemo-radiotherapy for stage III disease. Here we also discuss the current status of predictive biomarkers for anti-EGFR mAbs when added to first-line CT in patients with advanced NSCLC. Finally, we focused on the relevance of EGFR fluorescence in situ hybridization (FISH)+ and immunohistochemistry (IHC)-Score ≥ 200 as predictive biomarkers for the selection of patients who would be most likely to derive a clinical benefit from treatment with CT in combination with anti-EGFR mAbs, with particular reference also to histology.
Collapse
Affiliation(s)
- Francesco Agustoni
- Division of Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Kenichi Suda
- Division of Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States; Division of Thoracic Surgery, Department of Surgery, Kindai University, Faculty of Medicine, Osaka-Sayama, Japan
| | - Hui Yu
- Division of Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Shengxiang Ren
- Division of Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States; Department of Medical Oncology, Shanghai Pulmonary Hospital and Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai, China
| | - Christopher J Rivard
- Division of Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Kim Ellison
- Division of Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Charles Caldwell
- Division of Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Leslie Rozeboom
- Division of Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Kristine Brovsky
- Division of Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Fred R Hirsch
- Division of Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States.
| |
Collapse
|
9
|
Randomized phase II trial of cixutumumab alone or with cetuximab for refractory recurrent/metastatic head and neck squamous cell carcinoma. Oral Oncol 2018; 82:83-90. [PMID: 29909907 DOI: 10.1016/j.oraloncology.2018.05.014] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2018] [Revised: 05/10/2018] [Accepted: 05/16/2018] [Indexed: 12/24/2022]
Abstract
OBJECTIVES Cixutumumab (CIX) and cetuximab (CET) monoclonal antibodies block ligand-binding to insulin-like growth factor-1 receptor (IGF-1R) and epidermal growth factor receptor (EGFR) respectively. The objective of this study was to assess the efficacy of CIX alone or combined with CET in recurrent/metastatic head and neck squamous cell carcinoma (R/M HNSCC) patients. METHODS In this open-label phase II trial, 91 R/M HNSCC patients who progressed within 90 days of platinum-based chemotherapy, were randomized to CIX 10 mg/kg alone or with CET 500 mg/m2 every 2 weeks. Patients were stratified by prior CET use. The primary endpoint was median progression-free survival (PFS). Exploratory biomarker assessments included relevant markers on archival tumor and serial cytokine/angiogenic-factor profiles in blood. RESULTS Forty-seven patients were treated with CIX monotherapy and 44 with combination. The median PFS was 1.9 and 2.0 months and clinical benefit rate (complete or partial responses and stable disease) was 5.9% and 15.3%, respectively. There was no exacerbation of CET toxicity by concurrent CIX exposure. Higher tumor expression of IGF-1 was associated with improved PFS in the CIX + CET arm while increased p-EGFR expression correlated with shorter PFS in patients receiving single agent CIX. Higher serum baseline levels of IGF-1 and IGFBP-3 correlated with improved PFS and overall survival (OS) in the CIX arm. Neither regimen resulted in improved PFS or OS compared to historical data with CET alone. CONCLUSION The results of this study do not support the use of cixutumumab alone or with cetuximab in unselected patients with R/M HNSCC.
Collapse
|
10
|
Argiris A, Lee JW, Stevenson J, Sulecki MG, Hugec V, Choong NW, Saltzman JN, Song W, Hansen RM, Evans TL, Ramalingam SS, Schiller JH. Phase II randomized trial of carboplatin, paclitaxel, bevacizumab with or without cixutumumab (IMC-A12) in patients with advanced non-squamous, non-small-cell lung cancer: a trial of the ECOG-ACRIN Cancer Research Group (E3508). Ann Oncol 2018; 28:3037-3043. [PMID: 28950351 DOI: 10.1093/annonc/mdx534] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Background Cixutumumab is a fully human IgG1 monoclonal antibody to the insulin-like growth factor type I receptor that can potentially reverse resistance and enhance the efficacy of chemotherapy. Methods Bevacizumab-eligible patients with stage IV or recurrent non-squamous, non-small-cell lung cancer and good performance status were randomized to receive standard doses of paclitaxel, carboplatin, and bevacizumab to a maximum of six cycles followed by bevacizumab maintenance (CPB) until progression (arm A) or CPB plus cixutumumab 6 mg/kg i.v. weekly (arm B). Results Of 175 patients randomized, 153 were eligible and treated (78 in arm A; 75 in arm B). The median progression-free survival was 5.8 months (95% CI 5.4-7.1) in arm A versus 7 months (95% CI 5.7-7.6) in arm B (P = 0.33); hazard ratio 0.92 (95% CI 0.65-1.31). Objective response was 46.2% versus 58.7% in arm A versus arm B (P = 0.15). The median overall survival was 16.2 months in arm A versus 16.1 months in arm B (P = 0.95). Grade 3/4 neutropenia and febrile neutropenia, thrombocytopenia, fatigue, and hyperglycemia were increased with cixutumumab. Conclusions The addition of cixutumumab to CPB increased toxicity without improving efficacy and is not recommended for further development in non-small-cell lung cancer. Both treatment groups had longer OS than historical controls which may be attributed to several factors, and emphasizes the value of a comparator arm in phase II trials. ClinicalTrials.gov Identifier NCT00955305.
Collapse
Affiliation(s)
- A Argiris
- Medical Oncology, Hygeia Hospital, Athens, Greece.,Medical Oncology, Thomas Jefferson University, Philadelphia
| | - J W Lee
- Dana-Farber Cancer Institute, ECOG-ACRIN Biostatistics Center, Boston
| | - J Stevenson
- Medical Oncology, Cleveland Clinic Foundation, Cleveland
| | - M G Sulecki
- Medical Oncology, University of Pittsburgh Cancer Institute, Pittsburgh
| | - V Hugec
- Medical Oncology, Minnesota Oncology, Minneapolis, Lake Elmo
| | | | - J N Saltzman
- Medical Oncology, University Hospitals Seidman Cancer Center, Case Western Reserve University, Cleveland
| | - W Song
- Pottstown Memor Reg Cancer Ctr, Pottstown
| | - R M Hansen
- Medical Oncology, Oconomowoc Memorial Hospital, Oconomowoc
| | - T L Evans
- Medical Oncology, University of Pennsylvania, Philadelphia
| | - S S Ramalingam
- The Winship Cancer Institute of Emory University, Atlanta
| | - J H Schiller
- Medical Oncology, The University of Texas Southwestern Medical Center, Dallas, USA
| |
Collapse
|
11
|
Update of IGF-1 receptor inhibitor (ganitumab, dalotuzumab, cixutumumab, teprotumumab and figitumumab) effects on cancer therapy. Oncotarget 2018; 8:29501-29518. [PMID: 28427155 PMCID: PMC5438747 DOI: 10.18632/oncotarget.15704] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Accepted: 02/06/2017] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Prognostic studies of insulin-like growth factor-1 receptor(IGF-1R) inhibitors in cancer therapy had promising results in infratests, which exhibited that IGF-1R signalling was crucial in cancer cells growth. However, the conclusion of later clinical trials revealed a dim future for IGF-1R inhibitors to treat cancer. We conducted this analysis to figure out how IGF-1R inhibitors acted in clinical cancer therapy. MATERIAL AND METHODS We searched up-to-date studies about the single agent of IGF-1R inhibitors or combination with other therapies in solid tumor. Five IGF-1R anti-agents were involved. The primary endpoint was progression-free survival (PFS). The secondary endpoint was overall survival (OS). RESULT 17studies were enrolled. The results was not significant in overall survival (I2=37.1%, P=0.080, HR=1.08, 95% CI=0.97-1.21) and in progression-free survival (I2=0.0%, P=0.637, HR=1.05, 95% CI=0.98-1.12). OS for dalotuzumab, breast cancer, colorectal cancer, and PFS for prostate cancer even indicated harmful effects. CONCLUSION So far, anti-IGF-1R mono-antibodies did not make significant differences in solid tumor prognosis. On the contrary, pessimistic effects were shown in the dalotuzumab, breast cancer, colorectal cancer and prostate cancer subgroups. Further studies of IGF-1R anti-agents were needed, but unwarranted in unselected patients by predictive biomarkers.
Collapse
|
12
|
Husain H, Scur M, Murtuza A, Bui N, Woodward B, Kurzrock R. Strategies to Overcome Bypass Mechanisms Mediating Clinical Resistance to EGFR Tyrosine Kinase Inhibition in Lung Cancer. Mol Cancer Ther 2017; 16:265-272. [PMID: 28159915 DOI: 10.1158/1535-7163.mct-16-0105] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Revised: 10/20/2016] [Accepted: 10/24/2016] [Indexed: 11/16/2022]
Abstract
The vast majority of patients with metastatic lung cancers who initially benefit from EGFR-targeted therapies eventually develop resistance. An increasing understanding of the number and complexity of resistance mechanisms highlights the challenge of treating tumors resistant to EGFR inhibitors. Resistance mechanisms include new, second-site mutations within EGFR (e.g., T790M and C797S), upregulation of MET kinase, upregulation of insulin growth factor receptor (IGFR), HER2 amplification, increased expression of AXL, BIM modulation, NF-κB activation, histologic switch to small-cell cancer, epithelial-to-mesenchymal transition, PDL1 expression with subsequent immune tolerance, and release of cytokines such as TGFβ and IL6. Herein, we review the growing body of knowledge regarding EGFR bypass pathways, and the development of new drugs and combination treatment strategies to overcome resistance. Mol Cancer Ther; 16(2); 265-72. ©2017 AACR.
Collapse
Affiliation(s)
- Hatim Husain
- Division of Hematology and Oncology, Center for Personalized Cancer Therapy, University of California, San Diego, Moores Cancer Center, La Jolla, California.
| | - Michael Scur
- Division of Hematology and Oncology, Center for Personalized Cancer Therapy, University of California, San Diego, Moores Cancer Center, La Jolla, California
| | - Ayesha Murtuza
- Division of Hematology and Oncology, Center for Personalized Cancer Therapy, University of California, San Diego, Moores Cancer Center, La Jolla, California
| | - Nam Bui
- Division of Hematology and Oncology, Center for Personalized Cancer Therapy, University of California, San Diego, Moores Cancer Center, La Jolla, California
| | - Brian Woodward
- Division of Hematology and Oncology, Center for Personalized Cancer Therapy, University of California, San Diego, Moores Cancer Center, La Jolla, California
| | - Razelle Kurzrock
- Division of Hematology and Oncology, Center for Personalized Cancer Therapy, University of California, San Diego, Moores Cancer Center, La Jolla, California.
| |
Collapse
|
13
|
Paradigm shift of therapeutic management of brain metastases in EGFR-mutant non-small cell lung cancer in the era of targeted therapy. Med Oncol 2017; 34:121. [PMID: 28555261 DOI: 10.1007/s12032-017-0978-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Accepted: 05/09/2017] [Indexed: 12/25/2022]
Abstract
Non-small cell lung cancer (NSCLC) patients with epidermal growth factor receptor (EGFR) mutations commonly present brain metastases (BM) at the time of NSCLC diagnosis or during the clinical course. Conventionally, the prognosis of BM has been extremely poor, but the advent of EGFR-tyrosine kinase inhibitors (TKIs) has drastically improved the prognosis in these patients. Despite the presence of the blood-brain barrier, EGFR-TKIs have dramatic therapeutic effects on both BM and extracranial disease. In addition, recent systemic chemotherapies reportedly play a role in controlling BM. These treatment modalities can potentially replace whole brain radiotherapy (WBRT) to prevent or delay neurocognitive decline. Therefore, how to utilize these treatments is one issue. The other issue is what kind of treatment is best for recurrence after TKI therapy. Recent reports have shown a positive effect of a combination therapy of EGFR-TKI and radiotherapy on BM. Although neurocognitive decline is underscored when WBRT is considered, a survival benefit from WBRT has been proven especially in the potential long survivors with good prognostic index, especially disease-specific graded prognostic index (DS-GPA). In this review, treatment strategy including chemotherapeutic agents and radiotherapy is discussed in terms of risk-benefit balance in conjunction with DS-GPA.
Collapse
|
14
|
Cao H, Cui L, Ma W, Zhu L, Wang K, Ni Y, Wang Y, Du J. Adverse Events and Efficacy of Cixutumumab in Phase II Clinical Trials: A systematic Review and Meta-Analysis. Clin Drug Investig 2017; 37:135-153. [PMID: 27858328 DOI: 10.1007/s40261-016-0475-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND AND OBJECTIVES Cixutumumab is a monoclonal antibody targeting insulin-like growth factor 1 receptor (IGF1R). We sought to evaluate the efficacy of cixutumumab in the treatment of cancer, and to comprehensively assess the associated adverse events in phase II clinical trials. METHODS Data were collected from PubMed, Embase, and Clinicaltrials.gov. The improvement on progression-free survival (PFS) was evaluated by hazard ratio (HR) and 95% confidence intervals (95% CIs). We also carried a meta-analysis to comprehensively evaluate the incidence of adverse events. RESULTS The adverse events that were mentioned most frequently were hyperglycemia, anemia, nausea, fatigue, and thrombocytopenia. The most frequent adverse events were hyponatremia (40.28%), fatigue (35.18%), and skin rash (35.11%). Results showed that cixutumumab treatments did not benefit PFS (HR 1.03, 95% CI 0.83-1.26, p = 0.979). The complete response (CR) was rarely seen in phase II trials. CONCLUSIONS Cixutumumab was well tolerated when used alone and in combination therapies, but its antitumor activity was low in the existing phase II clinical trials. An acceptable incidence of adverse effects supports further investigation of this drug, provided that it shows antitumor activity in combination with other drugs.
Collapse
Affiliation(s)
- Hongxin Cao
- Institute of Oncology, Shandong Provincial Hospital Affiliated to Shandong University, Shandong University, Jinan, 250021, China.,Department of Chemotherapy, Cancer Center, Qilu Hospital of Shandong University, 8, Jinan, Shandong, 250012, People's Republic of China
| | - Lixuan Cui
- Institute of Oncology, Shandong Provincial Hospital Affiliated to Shandong University, Shandong University, Jinan, 250021, China
| | - Wei Ma
- Department of Thoracic Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Shandong University, Jinan, 250021, China
| | - Linhai Zhu
- Department of Thoracic Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Shandong University, Jinan, 250021, China
| | - Kai Wang
- Institute of Oncology, Shandong Provincial Hospital Affiliated to Shandong University, Shandong University, Jinan, 250021, China
| | - Yang Ni
- Department of Oncology, Shandong Provincial Hospital Affiliated to Shandong University, Shandong University, Jinan, 250021, China
| | - Yibing Wang
- Department of Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, 250021, China.
| | - Jiajun Du
- Institute of Oncology, Shandong Provincial Hospital Affiliated to Shandong University, Shandong University, Jinan, 250021, China. .,Department of Thoracic Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Shandong University, Jinan, 250021, China.
| |
Collapse
|
15
|
Li DL, Chen XR, Ma SX, Chen LK. Long-lasting response to third-line crizotinib treatment in a patient with non-small cell lung cancer with brain metastases and poor performance status. Thorac Cancer 2016; 7:619-622. [PMID: 27766783 PMCID: PMC5193012 DOI: 10.1111/1759-7714.12383] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Revised: 07/05/2016] [Accepted: 07/05/2016] [Indexed: 11/30/2022] Open
Abstract
Identification of the anaplastic lymphoma kinase (ALK) gene has refined the classification of non‐small cell lung cancer (NSCLC) and promoted research on molecularly targeted drugs such as crizotinib, an ALK inhibitor with good efficacy, in ALK‐rearranged NSCLC. At present, few studies have reported the efficacy of crizotinib in patients with ALK‐rearranged NSCLC with brain metastases. In a patient with NSCLC harboring ALK‐rearrangement who had brain metastases and poor performance status (PS), we obtained a durable response with crizotinib administered following multi‐line chemotherapy regimens.
Collapse
Affiliation(s)
- De-Lan Li
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, and Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Xin-Ru Chen
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, and Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Shu-Xiang Ma
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, and Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Li-Kun Chen
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, and Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
| |
Collapse
|
16
|
Sclafani F, Kim TY, Cunningham D, Kim TW, Tabernero J, Schmoll HJ, Roh JK, Kim SY, Park YS, Guren TK, Hawkes E, Clarke SJ, Ferry D, Frodin JE, Ayers M, Nebozhyn M, Peckitt C, Loboda A, Watkins DJ. Dalotuzumab in chemorefractory KRAS exon 2 mutant colorectal cancer: Results from a randomised phase II/III trial. Int J Cancer 2016; 140:431-439. [PMID: 27681944 DOI: 10.1002/ijc.30453] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2016] [Revised: 07/15/2016] [Accepted: 08/19/2016] [Indexed: 12/30/2022]
Abstract
Limited data are available on the efficacy of anti-IGF-1R agents in KRAS mutant colorectal cancer (CRC). We analysed the outcome of 69 chemorefractory, KRAS exon 2 mutant CRC patients who were enrolled in a double-blind, randomised, phase II/III study of irinotecan and cetuximab plus dalotuzumab 10 mg/kg once weekly (arm A), dalotuzumab 7.5 mg/kg every second week (arm B) or placebo (arm C). Objective response rate (5.6% vs. 3.1% vs. 4.8%), median progression-free survival (2.7 vs. 2.6 vs. 1.4 months) and overall survival (7.8 vs. 10.3 vs. 7.8 months) were not statistically significantly different between treatment groups. Most common grade ≥3 treatment-related toxicities included neutropenia, diarrhoea, hyperglycaemia, fatigue and dermatitis acneiform. Expression of IGF-1R, IGF-1, IGF-2 and EREG by quantitative real-time polymerase chain reaction was assessed in 351 patients from the same study with available data on KRAS exon 2 mutational status. Median cycle threshold values for all biomarkers were significantly lower (i.e., higher expression, p < 0.05) among patients with KRAS wild-type compared to those with KRAS exon 2 mutant tumours. No significant changes were found according to location of the primary tumour with only a trend towards lower expression of IGF-1 in colon compared to rectal cancers (p = 0.06). Albeit limited by the small sample size, this study does not appear to support a potential role for anti-IGF-1R agents in KRAS exon 2 mutant CRC. Data on IGF-1R, IGF-1 and IGF-2 expression here reported may be useful for patient stratification in future trials with inhibitors of the IGF pathway.
Collapse
Affiliation(s)
- Francesco Sclafani
- The Royal Marsden NHS Foundation Trust, London and Surrey, United Kingdom
| | - Tae Y Kim
- Seoul National University College of Medicine, Seoul, Korea
| | - David Cunningham
- The Royal Marsden NHS Foundation Trust, London and Surrey, United Kingdom
| | - Tae W Kim
- Department of Oncology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Josep Tabernero
- Vall d'Hebron University Hospital and Institute of Oncology (VHIO), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Hans J Schmoll
- Department of Internal Medicine, University Clinic Halle (Saale), Martin Luther University Halle-Wittenberg, Halle, Germany
| | - Jae K Roh
- College of Medicine, Yonsey Cancer Center, Yonsey University, Seoul, Korea
| | - Sun Y Kim
- Center for Colorectal Cancer, National Cancer Center, Seoul, Korea
| | - Young S Park
- Department of Medicine, Division of Hematology/Oncology, Samsung Medical Center, Seoul, Korea
| | - Tormod K Guren
- Department of Oncology and K.G. Jebsen Colorectal Cancer Research Centre, Oslo University Hospital, Oslo, Norway
| | - Eliza Hawkes
- The Royal Marsden NHS Foundation Trust, London and Surrey, United Kingdom
| | - Stephen J Clarke
- Concord Repatriation General Hospital, Concord, Sydney, Australia
| | - David Ferry
- New Cross Hospital, Wolverhamptom, United Kingdom
| | | | | | | | - Clare Peckitt
- The Royal Marsden NHS Foundation Trust, London and Surrey, United Kingdom
| | | | - David J Watkins
- The Royal Marsden NHS Foundation Trust, London and Surrey, United Kingdom
| |
Collapse
|
17
|
Khalifa J, Amini A, Popat S, Gaspar LE, Faivre-Finn C. Brain Metastases from NSCLC: Radiation Therapy in the Era of Targeted Therapies. J Thorac Oncol 2016; 11:1627-43. [PMID: 27343440 DOI: 10.1016/j.jtho.2016.06.002] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Revised: 06/02/2016] [Accepted: 06/09/2016] [Indexed: 02/07/2023]
Abstract
Brain metastases (BMs) will develop in a large proportion of patients with NSCLC throughout the course of their disease. Among patients with NSCLC with oncogenic drivers, mainly EGFR activating mutations and anaplastic lymphoma receptor tyrosine kinase gene (ALK) rearrangements, the presence of BM is a common secondary localization of disease both at the time of diagnosis and at relapse. Because of the limited penetration of a wide range of drugs across the blood-brain barrier, radiotherapy is considered the cornerstone of treatment of BMs. However, evidence of dramatic intracranial response rates has been reported in recent years with targeted therapies such as tyrosine kinase inhibitors and has been supported by new insights into pharmacokinetics to increase rates of tyrosine kinase inhibitors' penetration of the cerebrospinal fluid (CSF). In this context, the combination of brain radiotherapy and targeted therapies seems relevant, and there is a strong radiobiological rationale to harness the radiosentizing effect of the drugs. Nevertheless, to date, there is a paucity of high-level clinical evidence supporting the combination of brain radiotherapy and targeted therapies in patients with NSCLC and BMs, and there are often methodological biases in reported studies, such as the lack of stratification by mutation status. Moreover, among asymptomatic patients not suitable for ablative treatment, this strategy is challenged by the promising results associated with the administration of targeted therapies alone. Herein, we review the biological rationale to combine targeted therapies and brain radiotherapy for patients with NSCLC and BMs, report the clinical data available to date, and discuss future directions to improve outcome in this group of patients.
Collapse
Affiliation(s)
- Jonathan Khalifa
- Radiotherapy Related Research, The Christie National Health Service Foundation Trust, Manchester, United Kingdom.
| | - Arya Amini
- Department of Radiation Oncology, University of Colorado School of Medicine, Aurora, Colorado
| | - Sanjay Popat
- Lung Cancer Unit, Royal Marsden Hospital, London, United Kingdom
| | - Laurie E Gaspar
- Department of Radiation Oncology, University of Colorado School of Medicine, Aurora, Colorado
| | - Corinne Faivre-Finn
- Radiotherapy Related Research, The Christie National Health Service Foundation Trust, Manchester, United Kingdom; Manchester Academic Health Science Centre, Institute of Cancer Sciences, Manchester Cancer Research Centre, The University of Manchester, Manchester, United Kingdom
| |
Collapse
|
18
|
Toyokawa G, Seto T, Takenoyama M, Ichinose Y. Insights into brain metastasis in patients with ALK+ lung cancer: is the brain truly a sanctuary? Cancer Metastasis Rev 2016; 34:797-805. [PMID: 26342831 PMCID: PMC4661196 DOI: 10.1007/s10555-015-9592-y] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Anaplastic lymphoma kinase (ALK) has been identified to exert a potent transforming activity through its rearrangement in non-small cell lung cancer (NSCLC), and patients (pts) with ALK rearrangement can be treated more successfully with ALK inhibitors, such as crizotinib, alectinib, and ceritinib, than with chemotherapy. Despite the excellent efficacy of ALK inhibitors, resistance to these drugs is inevitably encountered in most ALK-rearranged pts. Cases of resistance are subtyped into three groups, i.e., systemic, oligo, and central nervous system (CNS) types, with the CNS being used to be considered a sanctuary. With regard to the management of CNS lesions in pts with ALK+ NSCLC, a growing body of evidence has gradually demonstrated the intracranial (IC) efficacy of ALK inhibitor (ALKi) in ALK+ NSCLC pts with brain metastases (BMs). Although the efficacy of crizotinib for the CNS lesions remains controversial, a recent retrospective investigation of ALK+ pts with BM enrolled in PROFILE 1005 and PROFILE 1007 demonstrated that crizotinib is associated with a high disease control rate for BM. However, BM comprises the most common site of progressive disease in pts with or without baseline BMs, which is a serious problem for crizotinib. Furthermore, alectinib can be used to achieve strong and long-lasting inhibitory effects on BM. In addition to alectinib, the IC efficacy of other next-generation ALK inhibitors, such as ceritinib, AP26113 and PF-06463922, has been demonstrated. In this article, we review the latest evidence regarding the BM and IC efficacy of ALK inhibitors in pts with ALK+ NSCLC.
Collapse
Affiliation(s)
- Gouji Toyokawa
- Department of Thoracic Oncology, National Kyushu Cancer Center, 3-1-1 Notame, Minami-ku, Fukuoka, 811-1395, Japan.
| | - Takashi Seto
- Department of Thoracic Oncology, National Kyushu Cancer Center, 3-1-1 Notame, Minami-ku, Fukuoka, 811-1395, Japan
| | - Mitsuhiro Takenoyama
- Department of Thoracic Oncology, National Kyushu Cancer Center, 3-1-1 Notame, Minami-ku, Fukuoka, 811-1395, Japan
| | - Yukito Ichinose
- Department of Thoracic Oncology, National Kyushu Cancer Center, 3-1-1 Notame, Minami-ku, Fukuoka, 811-1395, Japan
| |
Collapse
|
19
|
Losanno T, Rossi A, Maione P, Napolitano A, Gridelli C. Anti-EGFR and antiangiogenic monoclonal antibodies in metastatic non-small-cell lung cancer. Expert Opin Biol Ther 2016; 16:747-58. [PMID: 26950292 DOI: 10.1517/14712598.2016.1163333] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION In recent years, several clinical trials have evaluated the efficacy and safety of biological therapies in lung cancer. Epidermal growth factor receptor (EGFR) and the axis vascular endothelial growth factor receptor (VEGF/VEGFR) are targeted by small molecules and monoclonal antibodies (mAbs), especially in non-squamous non-small-cell lung cancer (NSCLC). AREAS COVERED The current state of the art of anti-EGFR and antiangiogenic monoclonal antibodies in metastatic NSCLC is reviewed and discussed. EXPERT OPINION Bevacizumab and cetuximab are the most studied mAbs in NSCLC, but only bevacizumab is in clinical practice in the first-line setting. Necitumumab is a new anti-EGFR monoclonal antibody that improves survival when combined to cisplatin/gemcitabine chemotherapy and has been approved in first-line advanced NSCLC. Ramucirumab, an antiangiogenic drug binding with high affinity to VEGFR-2, improves the results of chemotherapy alone when administered with docetaxel and has been approved in second-line setting. Moreover, the novel combination of bevacizumab and erlotinib is very promising for the treatment of patients with NSCLC harbouring EGFR mutations. The association of antiangiogenic mAbs and immunotherapy is under investigation too.
Collapse
Affiliation(s)
- Tania Losanno
- a Department of Experimental Medicine , University 'Sapienza' , Rome , Italy
| | - Antonio Rossi
- b Division of Medical Oncology , S.G. Moscati Hospital , Avellino , Italy
| | - Paolo Maione
- b Division of Medical Oncology , S.G. Moscati Hospital , Avellino , Italy
| | - Alba Napolitano
- c Division of Pharmacy , S.G. Moscati Hospital , Avellino , Italy
| | - Cesare Gridelli
- b Division of Medical Oncology , S.G. Moscati Hospital , Avellino , Italy
| |
Collapse
|
20
|
Nurwidya F, Andarini S, Takahashi F, Syahruddin E, Takahashi K. Implications of Insulin-like Growth Factor 1 Receptor Activation in Lung Cancer. Malays J Med Sci 2016; 23:9-21. [PMID: 27418865 PMCID: PMC4934714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 04/14/2016] [Indexed: 06/06/2023] Open
Abstract
Insulin-like growth factor 1 receptor (IGF1R) has been intensively investigated in many preclinical studies using cell lines and animal models, and the results have provided important knowledge to help improve the understanding of cancer biology. IGF1R is highly expressed in patients with lung cancer, and high levels of circulating insulin-like growth factor 1 (IGF1), the main ligand for IGF1R, increases the risk of developing lung malignancy in the future. Several phase I clinical trials have supported the potential use of an IGF1R-targeted strategy for cancer, including lung cancer. However, the negative results from phase III studies need further attention, especially in selecting patients with specific molecular signatures, who will gain benefits from IGF1R inhibitors with minimal side effects. This review will discuss the basic concept of IGF1R in lung cancer biology, such as epithelial-mesenchymal transition (EMT) induction and cancer stem cell (CSC) maintenance, and also the clinical implications of IGF1R for lung cancer patients, such as prognostic value and cancer therapy resistance.
Collapse
Affiliation(s)
- Fariz Nurwidya
- Department of Pulmonology and Respiratory Medicine, University of Indonesia Faculty of Medicine, Persahabatan Hospital, Jalan Persahabatan Raya No.1, Jakarta 13230, Indonesia
- Department of Respiratory Medicine, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Sita Andarini
- Department of Pulmonology and Respiratory Medicine, University of Indonesia Faculty of Medicine, Persahabatan Hospital, Jalan Persahabatan Raya No.1, Jakarta 13230, Indonesia
| | - Fumiyuki Takahashi
- Department of Respiratory Medicine, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Elisna Syahruddin
- Department of Pulmonology and Respiratory Medicine, University of Indonesia Faculty of Medicine, Persahabatan Hospital, Jalan Persahabatan Raya No.1, Jakarta 13230, Indonesia
| | - Kazuhisa Takahashi
- Department of Respiratory Medicine, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
| |
Collapse
|
21
|
Sechler M, Cizmic AD, Avasarala S, Van Scoyk M, Brzezinski C, Kelley N, Bikkavilli RK, Winn RA. Non-small-cell lung cancer: molecular targeted therapy and personalized medicine - drug resistance, mechanisms, and strategies. PHARMACOGENOMICS & PERSONALIZED MEDICINE 2013; 6:25-36. [PMID: 23690695 PMCID: PMC3656464 DOI: 10.2147/pgpm.s26058] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Targeted therapies for cancer bring the hope of specific treatment, providing high efficacy and in some cases lower toxicity than conventional treatment. Although targeted therapeutics have helped immensely in the treatment of several cancers, like chronic myelogenous leukemia, colon cancer, and breast cancer, the benefit of these agents in the treatment of lung cancer remains limited, in part due to the development of drug resistance. In this review, we discuss the mechanisms of drug resistance and the current strategies used to treat lung cancer. A better understanding of these drug-resistance mechanisms could potentially benefit from the development of a more robust personalized medicine approach for the treatment of lung cancer.
Collapse
Affiliation(s)
- Marybeth Sechler
- Division of Pulmonary Sciences and Critical Care, University of Colorado, Aurora, CO, USA ; Program in Cancer Biology, University of Colorado, Aurora, CO, USA
| | | | | | | | | | | | | | | |
Collapse
|