201
|
Chen AP, Kummar S, Moore N, Rubinstein LV, Zhao Y, Williams PM, Palmisano A, Sims D, O'Sullivan Coyne G, Rosenberger CL, Simpson M, Raghav KPS, Meric-Bernstam F, Leong S, Waqar S, Foster JC, Konaté MM, Das B, Karlovich C, Lih CJ, Polley E, Simon R, Li MC, Piekarz R, Doroshow JH. Molecular Profiling-Based Assignment of Cancer Therapy (NCI-MPACT): A Randomized Multicenter Phase II Trial. JCO Precis Oncol 2021; 5:PO.20.00372. [PMID: 33928209 PMCID: PMC8078898 DOI: 10.1200/po.20.00372] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 11/10/2020] [Accepted: 11/24/2020] [Indexed: 12/19/2022] Open
Abstract
This trial assessed the utility of applying tumor DNA sequencing to treatment selection for patients with advanced, refractory cancer and somatic mutations in one of four signaling pathways by comparing the efficacy of four study regimens that were either matched to the patient's aberrant pathway (experimental arm) or not matched to that pathway (control arm). MATERIALS AND METHODS Adult patients with an actionable mutation of interest were randomly assigned 2:1 to receive either (1) a study regimen identified to target the aberrant pathway found in their tumor (veliparib with temozolomide or adavosertib with carboplatin [DNA repair pathway], everolimus [PI3K pathway], or trametinib [RAS/RAF/MEK pathway]), or (2) one of the same four regimens, but chosen from among those not targeting that pathway. RESULTS Among 49 patients treated in the experimental arm, the objective response rate was 2% (95% CI, 0% to 10.9%). One of 20 patients (5%) in the experimental trametinib cohort had a partial response. There were no responses in the other cohorts. Although patients and physicians were blinded to the sequencing and random assignment results, a higher pretreatment dropout rate was observed in the control arm (22%) compared with the experimental arm (6%; P = .038), suggesting that some patients may have had prior tumor mutation profiling performed that led to a lack of participation in the control arm. CONCLUSION Further investigation, better annotation of predictive biomarkers, and the development of more effective agents are necessary to inform treatment decisions in an era of precision cancer medicine. Increasing prevalence of tumor mutation profiling and preference for targeted therapy make it difficult to use a randomized phase II design to evaluate targeted therapy efficacy in an advanced disease setting.
Collapse
Affiliation(s)
- Alice P. Chen
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD
| | - Shivaani Kummar
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD
- Knight Cancer Institute, Oregon Health and Science University, Portland, OR
| | - Nancy Moore
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD
| | | | - Yingdong Zhao
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD
| | - P. Mickey Williams
- Molecular Characterization Laboratory, Frederick National Laboratory for Cancer Research, Frederick, MD
| | - Alida Palmisano
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD
- General Dynamics Information Technology (GDIT), Falls Church, VA
| | - David Sims
- Molecular Characterization Laboratory, Frederick National Laboratory for Cancer Research, Frederick, MD
| | | | | | - Mel Simpson
- Applied/Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick, MD
| | - Kanwal P. S. Raghav
- Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Funda Meric-Bernstam
- Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX
| | | | - Saiama Waqar
- Department of Medical Oncology, Washington University School of Medicine, St Louis, MO
| | - Jared C. Foster
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD
| | - Mariam M. Konaté
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD
| | - Biswajit Das
- Molecular Characterization Laboratory, Frederick National Laboratory for Cancer Research, Frederick, MD
| | - Chris Karlovich
- Molecular Characterization Laboratory, Frederick National Laboratory for Cancer Research, Frederick, MD
| | - Chih-Jian Lih
- Molecular Characterization Laboratory, Frederick National Laboratory for Cancer Research, Frederick, MD
| | - Eric Polley
- Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN
| | - Richard Simon
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD
| | - Ming-Chung Li
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD
| | - Richard Piekarz
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD
| | - James H. Doroshow
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD
- Center for Cancer Research, National Cancer Institute, Bethesda, MD
| |
Collapse
|
202
|
Jeong H, Jeong JH, Kim KP, Lee SS, Oh DW, Park DH, Song TJ, Park Y, Hong SM, Ryoo BY, Yoo C. Feasibility of HER2-Targeted Therapy in Advanced Biliary Tract Cancer: A Prospective Pilot Study of Trastuzumab Biosimilar in Combination with Gemcitabine Plus Cisplatin. Cancers (Basel) 2021; 13:cancers13020161. [PMID: 33418871 PMCID: PMC7825072 DOI: 10.3390/cancers13020161] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 12/24/2020] [Accepted: 01/02/2021] [Indexed: 12/22/2022] Open
Abstract
The prognosis of advanced biliary tract cancer (BTC) is poor with the standard gemcitabine and cisplatin (GemCis) regimen. Given that the rates of human epidermal growth factor receptor 2 (HER2) positivity in BTC reaches around 15%, HER2-targeted therapy needs further investigation. This study aims to evaluate the preliminary efficacy/safety of first-line trastuzumab-pkrb plus GemCis in patients with advanced BTC. Patients with unresectable/metastatic HER2-positive BTC received trastuzumab-pkrb (on day 1 of each cycle, 8 mg/kg for the first cycle and 6 mg/kg for subsequent cycles), gemcitabine (1000 mg/m2 on day 1 and 8) and cisplatin (25 mg/m2 on day 1 and 8) every 3 weeks. Of the 41 patients screened, 7 had HER2-positive tumours and 4 were enrolled. The median age was 72.5 years (one male). Primary tumour locations included extrahepatic (N = 2) and intrahepatic (N = 1) bile ducts, and gallbladder (N = 1). Best overall response was a partial response in two patients and stable disease in two patients. Median progression-free survival (PFS) was 6.1 months and median overall survival (OS) was not reached. The most common grade 3 adverse event was neutropenia (75%), but febrile neutropenia did not occur. No patient discontinued treatment due to adverse events. Trastuzumab-pkrb with GemCis showed promising preliminary feasibility in patients with HER2-positive advanced BTC.
Collapse
Affiliation(s)
- Hyehyun Jeong
- Asan Medical Center, Department of Oncology, University of Ulsan College of Medicine, Seoul 05505, Korea; (H.J.); (J.H.J.); (K.-P.K.); (B.-Y.R.)
| | - Jae Ho Jeong
- Asan Medical Center, Department of Oncology, University of Ulsan College of Medicine, Seoul 05505, Korea; (H.J.); (J.H.J.); (K.-P.K.); (B.-Y.R.)
| | - Kyu-Pyo Kim
- Asan Medical Center, Department of Oncology, University of Ulsan College of Medicine, Seoul 05505, Korea; (H.J.); (J.H.J.); (K.-P.K.); (B.-Y.R.)
| | - Sang Soo Lee
- Asan Medical Center, Department of Gastroenterology, University of Ulsan College of Medicine, Seoul 05505, Korea; (S.S.L.); (D.W.O.); (D.H.P.); (T.J.S.)
| | - Dong Wook Oh
- Asan Medical Center, Department of Gastroenterology, University of Ulsan College of Medicine, Seoul 05505, Korea; (S.S.L.); (D.W.O.); (D.H.P.); (T.J.S.)
| | - Do Hyun Park
- Asan Medical Center, Department of Gastroenterology, University of Ulsan College of Medicine, Seoul 05505, Korea; (S.S.L.); (D.W.O.); (D.H.P.); (T.J.S.)
| | - Tae Jun Song
- Asan Medical Center, Department of Gastroenterology, University of Ulsan College of Medicine, Seoul 05505, Korea; (S.S.L.); (D.W.O.); (D.H.P.); (T.J.S.)
| | - Yangsoon Park
- Asan Medical Center, Department of Pathology, University of Ulsan College of Medicine, Seoul 05505, Korea; (Y.P.); (S.-M.H.)
| | - Seung-Mo Hong
- Asan Medical Center, Department of Pathology, University of Ulsan College of Medicine, Seoul 05505, Korea; (Y.P.); (S.-M.H.)
| | - Baek-Yeol Ryoo
- Asan Medical Center, Department of Oncology, University of Ulsan College of Medicine, Seoul 05505, Korea; (H.J.); (J.H.J.); (K.-P.K.); (B.-Y.R.)
| | - Changhoon Yoo
- Asan Medical Center, Department of Oncology, University of Ulsan College of Medicine, Seoul 05505, Korea; (H.J.); (J.H.J.); (K.-P.K.); (B.-Y.R.)
- Correspondence: ; Tel.: +82-2-3010-1727
| |
Collapse
|
203
|
Vogel A, Saborowski A. Current and Future Systemic Therapies in Biliary Tract Cancer. Visc Med 2021; 37:32-38. [PMID: 33708816 DOI: 10.1159/000513969] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Accepted: 12/21/2020] [Indexed: 12/15/2022] Open
Abstract
Background Despite an increasing incidence, biliary tumors are still considered a rare tumor entity. Due to an often long clinically inapparent course and a lack of early detection strategies, surgical resection is often not possible at the time of diagnosis. Since 2010, chemotherapy with gemcitabine and cisplatin is considered the standard of care in the palliative situation. Only recently, first studies have been published or initiated that expand the treatment options in the first line and, for the first time, also suggest valid systemic approaches in the second line. Summary Molecularly targeted therapies in selected patient subgroups are rapidly changing the field. In addition to IDH1 mutations and FGFR2 fusions in patients with intrahepatic tumors, the therapeutic relevance of rare but targetable alterations such as HER2/neu amplification, NTRK fusions, or BRAF mutations should be considered in patients with biliary tract cancers. Key Message The current study landscape clearly shows that precision medicine will play an important role in the therapy of biliary malignancies and underlines the importance of early tumor genetic diagnostics. In this article we provide an overview of systemic therapy concepts in the adjuvant and palliative setting.
Collapse
Affiliation(s)
- Arndt Vogel
- Department of Gastroenterology, Hepatology and Endocrinology, Medical School Hannover, Hannover, Germany
| | - Anna Saborowski
- Department of Gastroenterology, Hepatology and Endocrinology, Medical School Hannover, Hannover, Germany
| |
Collapse
|
204
|
Feng F, Wu X, Shi X, Gao Q, Wu Y, Yu Y, Cheng Q, Li B, Yi B, Liu C, Hao Q, Zhang L, Gao C, Jiang X. Comprehensive analysis of genomic alterations of Chinese hilar cholangiocarcinoma patients. Int J Clin Oncol 2021; 26:717-727. [PMID: 33387086 DOI: 10.1007/s10147-020-01846-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 11/23/2020] [Indexed: 12/20/2022]
Abstract
BACKGROUND Cholangiocarcinoma (CCA) is a rare malignant tumor of the biliary system. The heterogeneity of CCA leads to the lack of effective targeted treatment for CCA subtypes. The molecular characteristic of hilar CCA (hCCA) is still unclear. METHODS A total of 63 hCCA patients were enrolled from Shanghai Eastern Hepatobiliary Surgery Hospital. Formalin-fixed, paraffin-embedded tumor tissues, and matched blood were collected and deep sequencing targeting 450 cancer genes were performed. Tumor mutation burden (TMB) was measured by an algorithm developed in-house. Correlation analysis was performed by Fisher's exact test. RESULTS The most commonly mutated genes were TP53 (51.7%), NF1 and KRAS (20%, for both), SMAD4 (16.7%), FAT3 and FRS2 (13.3%, for both), NF1 (11.7%), and KMT2C, MDM2, and ATM (10%, for each) in hCCA. ARID1A, GATA6, and PREX2 mutations commonly occurred in female and KMT2C mutations mainly occurred in patients under 60 years old. Statistical analysis showed the association between ARID1A mutation and tumor stage (P = 0.041) and between NF1 mutation and high TMB (P = 0.0095). Furthermore, ARID1B mutation was identified to associate with the poor prognosis of Chinese hCCA patients (P = 0.004). CONCLUSION The mutational characterization of hCCA is different from both extrahepatic CCA and intrahepatic CCA. ARID1B is a potential biomarker for prognosis prediction of Chinese hCCA patients.
Collapse
Affiliation(s)
- Feiling Feng
- Department of Biliary Tract I, Eastern Hepatobiliary Surgery Hospital, No.225, Changhai Road, Yangpu District, Shanghai, 200433, People's Republic of China
| | - Xiaobing Wu
- Department of Biliary Tract I, Eastern Hepatobiliary Surgery Hospital, No.225, Changhai Road, Yangpu District, Shanghai, 200433, People's Republic of China
| | - Xiaoliang Shi
- Origimed Co. Ltd, Shanghai, 201114, People's Republic of China
| | - Qingxiang Gao
- Department of Biliary Tract I, Eastern Hepatobiliary Surgery Hospital, No.225, Changhai Road, Yangpu District, Shanghai, 200433, People's Republic of China
| | - Yue Wu
- Department of Biliary Tract I, Eastern Hepatobiliary Surgery Hospital, No.225, Changhai Road, Yangpu District, Shanghai, 200433, People's Republic of China
| | - Yong Yu
- Department of Biliary Tract I, Eastern Hepatobiliary Surgery Hospital, No.225, Changhai Road, Yangpu District, Shanghai, 200433, People's Republic of China
| | - Qingbao Cheng
- Department of Biliary Tract I, Eastern Hepatobiliary Surgery Hospital, No.225, Changhai Road, Yangpu District, Shanghai, 200433, People's Republic of China
| | - Bin Li
- Department of Biliary Tract I, Eastern Hepatobiliary Surgery Hospital, No.225, Changhai Road, Yangpu District, Shanghai, 200433, People's Republic of China
| | - Bin Yi
- Department of Biliary Tract I, Eastern Hepatobiliary Surgery Hospital, No.225, Changhai Road, Yangpu District, Shanghai, 200433, People's Republic of China
| | - Chen Liu
- Department of Biliary Tract I, Eastern Hepatobiliary Surgery Hospital, No.225, Changhai Road, Yangpu District, Shanghai, 200433, People's Republic of China
| | - Qing Hao
- Origimed Co. Ltd, Shanghai, 201114, People's Republic of China
| | - Lin Zhang
- Origimed Co. Ltd, Shanghai, 201114, People's Republic of China
| | - Chunfang Gao
- Department of Biliary Tract I, Eastern Hepatobiliary Surgery Hospital, No.225, Changhai Road, Yangpu District, Shanghai, 200433, People's Republic of China.
| | - Xiaoqing Jiang
- Department of Biliary Tract I, Eastern Hepatobiliary Surgery Hospital, No.225, Changhai Road, Yangpu District, Shanghai, 200433, People's Republic of China.
| |
Collapse
|
205
|
Alyateem G, Nilubol N. Current Status and Future Targeted Therapy in Adrenocortical Cancer. Front Endocrinol (Lausanne) 2021; 12:613248. [PMID: 33732213 PMCID: PMC7957049 DOI: 10.3389/fendo.2021.613248] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 01/25/2021] [Indexed: 12/11/2022] Open
Abstract
Adrenocortical carcinoma (ACC) is a rare malignancy with a poor prognosis. The current treatment standards include complete surgical resection for localized resectable disease and systemic therapy with mitotane alone or in combination with etoposide, doxorubicin, and cisplatin in patients with advanced ACC. However, the efficacy of systemic therapy in ACC is very limited, with high rates of toxicities. The understanding of altered molecular pathways is critically important to identify effective treatment options that currently do not exist. In this review, we discuss the results of recent advanced in molecular profiling of ACC with the focus on dysregulated pathways from various genomic and epigenetic dysregulation. We discuss the potential translational therapeutic implication of molecular alterations. In addition, we review and summarize the results of recent clinical trials and ongoing trials.
Collapse
|
206
|
Adashek JJ, Subbiah V, Kurzrock R. From Tissue-Agnostic to N-of-One Therapies: (R)Evolution of the Precision Paradigm. Trends Cancer 2021; 7:15-28. [DOI: 10.1016/j.trecan.2020.08.009] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 07/29/2020] [Accepted: 08/27/2020] [Indexed: 12/14/2022]
|
207
|
Lengyel CG, Habeeb B, Khan SZ, El Bairi K, Altuna SC, Hussain S, Mazher SA, Trapani D, Petrillo A. Role of Her-2 in Gastrointestinal Tumours beyond Gastric Cancer: A Tool for Precision Medicine. GASTROINTESTINAL DISORDERS 2020; 3:1-22. [DOI: 10.3390/gidisord3010001] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Gastrointestinal (GI) tumors account for a quarter of all the cancer burden and a third of the global cancer-related mortality. Among them, some cancers retain a dismal prognosis; therefore, newer and innovative therapies are urgently needed in priority disease areas of high-unmet medical need. In this context, HER2 could be a relevant prognostic and predictive biomarker acting as a target for specific drugs. However, if the role of HER2 has been object of investigation for several years in gastric cancer, it is not well established in other GI malignancies. The aim of this narrative review was to portray the current landscape of the potential role of HER2 as a predictive biomarker for GI tumors beyond gastric cancer. In colon cancer, the benefit from anti-HER2 therapies is less clear than in gastric neoplasms for the lack of controlled studies. Pancreatic, biliary tract adenocarcinomas and hepatocarcinoma may derive a less clear clinical benefit by using anti-HER2 agents in HER2 positive tumors. Overall, the results are promising and seem to suggest that the integration of multiple modalities of therapies can optimize the cancer care. However, further prospective trials are needed to validate the use of personalized targeted therapies in this field.
Collapse
Affiliation(s)
- Csongor G. Lengyel
- Head and Neck Surgery, National Institute of Oncology, 1122 Budapest, Hungary
| | - Baker Habeeb
- Medical Oncology Department, Shaqlawa Teaching Hospital, Erbil 44001, Iraq
| | - Shah Z. Khan
- Department of Clinical Oncology, BINOR Cancer Hospital, Bannu 28100, Pakistan
| | | | | | - Sadaqat Hussain
- Northwest Cancer Center, Western Health and Social Care Trust, Altnagelvin Hospital, Londonderry BT47 6SB, UK
| | - Syed Ayub Mazher
- UT Southwestern Clements University Hospital, Dallas, TX 75390, USA
| | - Dario Trapani
- European Institute of Oncology, IRCCS, 20141 Milan, Italy
| | - Angelica Petrillo
- Medical Oncology Unit, Ospedale del Mare, 80147 Naples, Italy
- Division of Medical Oncology, Department of Precision Medicine, School of Medicine, University of Study of Campania “L.Vanvitelli”, 81100 Caserta, Italy
| |
Collapse
|
208
|
Collins A, Miles GJ, Powley IR, Hew R, Pringle JH, MacFarlane M, Pritchard C, Moss EL. Development of a patient-derived explant model for prediction of drug responses in endometrial cancer. Gynecol Oncol 2020; 160:557-567. [PMID: 33309417 DOI: 10.1016/j.ygyno.2020.11.033] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 11/27/2020] [Indexed: 12/15/2022]
Abstract
OBJECTIVE To undertake a pilot study to develop a novel Patient-Derived-Explant (PDE) model system for use in endometrial cancer (EC) that is capable of monitoring differential drug responses in a pre-clinical setting. METHODS Fresh tumour was obtained post-hysterectomy from 27 patients with EC. Tumours were cut into 1-3 mm3 explants that were cultured at the air-liquid interface for 16-24 h in culture media. Explants were cultured in different media conditions to optimise viability. Explants were also treated with carboplatin/paclitaxel or pembrolizumab for 24 h and processed into histology slides. Multiplexed immunofluorescence for Ki67 (proliferation marker), cPARP (apoptosis marker) and CAM 5.2 (tumour mask) was performed followed by image analysis and quantitation of biomarker expression. RESULTS EC samples are amenable to PDE culture with preserved histological architecture and PDE viability for up to 48 h, with the addition of autologous serum in culture media facilitating EC-PDE viability. Our PDE platform provides evidence of differential drug-response to conventional chemotherapeutics and immune checkpoint inhibition, and these responses can be assessed in the context of a preserved tumour microenvironment. CONCLUSIONS Our PDE platform represents a rapid, low-cost pre-clinical model which can be easily integrated into drug development pipelines. PDE culture preserves original tumour architecture and enables evaluation of spatial relationships in the tumour microenvironment. PDE culture has the potential for personalised drug-testing in a pre-clinical setting which is increasingly important in an era of personalised medicine in the treatment of EC.
Collapse
Affiliation(s)
- Anna Collins
- Leicester Cancer Research Centre, University of Leicester, Clinical Sciences Building, Leicester LE2 7LX, UK
| | - Gareth J Miles
- Leicester Cancer Research Centre, University of Leicester, Clinical Sciences Building, Leicester LE2 7LX, UK
| | - Ian R Powley
- Leicester Cancer Research Centre, University of Leicester, Clinical Sciences Building, Leicester LE2 7LX, UK
| | - Roger Hew
- Department of Pathology, University Hospitals of Leicester NHS Trust, Leicester Royal Infirmary, Sandringham Building, Leicester LE1 5WW, UK
| | - J Howard Pringle
- Leicester Cancer Research Centre, University of Leicester, Clinical Sciences Building, Leicester LE2 7LX, UK
| | - Marion MacFarlane
- MRC Toxicology Unit, Hodgkin Building, Lancaster Road, Leicester LE1 9HN, UK
| | - Catrin Pritchard
- Leicester Cancer Research Centre, University of Leicester, Clinical Sciences Building, Leicester LE2 7LX, UK.
| | - Esther L Moss
- Leicester Cancer Research Centre, University of Leicester, Clinical Sciences Building, Leicester LE2 7LX, UK.
| |
Collapse
|
209
|
Wang L, Cao H, Jiang C, He W, You Y, Peng K, Jin Y, Xia L. Previous Use of Anti-Vascular Endothelial Growth Factor Receptor Agents Decreases Efficacy of Fruquintinib in Metastatic Colorectal Cancer Refractory to Standard Therapies. Front Oncol 2020; 10:587692. [PMID: 33282739 PMCID: PMC7691567 DOI: 10.3389/fonc.2020.587692] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 10/19/2020] [Indexed: 12/31/2022] Open
Abstract
Purpose Fruquintinib is an anti-vascular endothelial growth factor receptor (VEGFR) agent. The FRESCO trial demonstrated that patients with metastatic colorectal cancer (mCRC) refractory to standard therapies could benefit from fruquintinib with tolerable adverse events (AEs). However, the efficacy and safety of fruquintinib in clinical practice has scarcely been reported, especially in patients with previous use of anti-VEGFR agents. Methods This retrospective study investigated the efficacy and safety of fruquintinib in patients with mCRC between January 2019 and December 2019. Progression-free survival (PFS) and overall survival (OS) were assessed by a Kaplan-Meier analysis and log-rank test. A Cox regression model was performed to identify independent prognostic factors. Results A total of 46 patients were included. The median PFS and OS were 3.1 months (95% confidence interval [CI], 1.9–4.3 months) and 9.0 months (95% CI, 7.2–10.8 months), respectively. Patients previously treated with anti-VEGFR agents had shorter median PFS compared with those without previous use of anti-VEGFR agents (1.9 vs. 3.7 months, P = 0.006), while the median OS was similar between the two groups (8.5 vs. 9.0 months, P = 0.992). Multivariate analysis revealed that the neutrophil-lymphocyte ratio (NLR) was an independent prognostic factor in PFS (hazard ratio [HR], 2.230; 95% CI, 1.191–4.517, P = 0.014) and OS (HR, 4.221; 95% CI, 1.683–10.586; P = 0.002). The most common non-hematological and hematological AEs were hand-foot syndrome (37.0%) and anemia (39.1%), respectively. Conclusion Fruquintinib was an effective third-line therapy in mCRC with tolerable AEs. Efficacy of fruquintinib was decreased in patients with previous use of anti-VEGFR agents. NLR was an independent prognostic factor in PFS and OS in patients treated with fruquintinib.
Collapse
|
210
|
Jin J, Riviere MK, Luo X, Dong Y. Bayesian methods for the analysis of early-phase oncology basket trials with information borrowing across cancer types. Stat Med 2020; 39:3459-3475. [PMID: 32717103 DOI: 10.1002/sim.8675] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Revised: 06/04/2020] [Accepted: 06/05/2020] [Indexed: 12/18/2022]
Abstract
Research in oncology has changed the focus from histological properties of tumors in a specific organ to a specific genomic aberration potentially shared by multiple cancer types. This motivates the basket trial, which assesses the efficacy of treatment simultaneously on multiple cancer types that have a common aberration. Although the assumption of homogeneous treatment effects seems reasonable given the shared aberration, in reality, the treatment effect may vary by cancer type, and potentially only a subgroup of the cancer types respond to the treatment. Various approaches have been proposed to increase the trial power by borrowing information across cancer types, which, however, tend to inflate the type I error rate. In this article, we review some representative Bayesian information borrowing methods for the analysis of early-phase basket trials. We then propose a novel method called the Bayesian hierarchical model with a correlated prior (CBHM), which conducts more flexible borrowing across cancer types according to sample similarity. We did simulation studies to compare CBHM with independent analysis and three information borrowing approaches: the conventional Bayesian hierarchical model, the EXNEX approach, and Liu's two-stage approach. Simulation results show that all information borrowing approaches substantially improve the power of independent analysis if a large proportion of the cancer types truly respond to the treatment. Our proposed CBHM approach shows an advantage over the existing information borrowing approaches, with a power similar to that of EXNEX or Liu's approach, but the potential to provide substantially better control of type I error rate.
Collapse
Affiliation(s)
- Jin Jin
- Department of Biostatistics, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, USA
| | - Marie-Karelle Riviere
- Department of Biostatistics and Programming, Research and Development, Sanofi, Chilly-Mazarin, France
| | - Xiaodong Luo
- Department of Biostatistics and Programming, Sanofi, Bridgewater, New Jersey, USA
| | - Yingwen Dong
- Department of Biostatistics and Programming Oncology, Sanofi, Cambridge, Massachusetts, USA
| |
Collapse
|
211
|
Meintker L, Haller F, Tögel L, Schmidt D, Waibel H, Hartmann A, Mackensen A, Meidenbauer N. Successful Targeting of BRAF V600E Mutation With Vemurafenib in a Treatment-Resistant Extragonadal Nonseminomatous Germ-Cell Tumor. JCO Precis Oncol 2020; 4:233-238. [PMID: 35050734 DOI: 10.1200/po.19.00377] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
- Lisa Meintker
- Department of Medicine 5 for Hematology and Oncology, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
| | - Florian Haller
- Institute of Pathology, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
| | - Lars Tögel
- Institute of Pathology, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
| | - Daniela Schmidt
- Clinic of Nuclear Medicine, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
| | - Heidi Waibel
- Department of Medicine 5 for Hematology and Oncology, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
| | - Arndt Hartmann
- Institute of Pathology, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
| | - Andreas Mackensen
- Department of Medicine 5 for Hematology and Oncology, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
| | - Norbert Meidenbauer
- Department of Medicine 5 for Hematology and Oncology, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
| |
Collapse
|
212
|
Basket trials: From tumour gnostic to tumour agnostic drug development. Cancer Treat Rev 2020; 90:102082. [DOI: 10.1016/j.ctrv.2020.102082] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 07/07/2020] [Accepted: 07/10/2020] [Indexed: 12/14/2022]
|
213
|
Fujii S, Magliocco AM, Kim J, Okamoto W, Kim JE, Sawada K, Nakamura Y, Kopetz S, Park WY, Tsuchihara K, Kim TW, Raghav K, Yoshino T. International Harmonization of Provisional Diagnostic Criteria for ERBB2-Amplified Metastatic Colorectal Cancer Allowing for Screening by Next-Generation Sequencing Panel. JCO Precis Oncol 2020; 4:6-19. [DOI: 10.1200/po.19.00154] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
PURPOSE ERBB2 amplification (human epidermal growth factor receptor 2 positivity [HER2+]) in metastatic colorectal cancer (mCRC) has important therapeutic implications that necessitate the need for accurate diagnostics. The study purpose was to establish and validate harmonized diagnostic criteria for HER2+ mCRC among 3 groups (GI-SCREEN-Japan, NCTN-SWOG-USA, and Korea). PATIENTS AND METHODS We assessed HER2 status by immunohistochemistry (IHC), ERBB2/CEP17 ratio, gene copy number (GCN) by fluorescence in situ hybridization (FISH), and copy number variation (CNV) using two targeted next-generation sequencing (NGS) panels. Tumor samples from 475 and 16 patients with mCRC in exploratory and validation cohorts, respectively, were used for cross-validation of the NGS panels. RESULTS Consensus diagnostic criteria among the 3 groups for HER2+ mCRC were established as follows: IHC 3+ or IHC 2+ and ERBB2/CEP17 ratio by FISH ≥ 2.0, tumor content > 10% for surgically resected specimens, and necessary tumor content not defined for biopsy specimens. The median GCN and CNV for HER2+ patients were 10.9 and 27.7 compared with 2.5 ( P < .0001) and 3.5 ( P < .0001), respectively, in HER2-negative patients. These findings were validated in a validation cohort (GCN, 16.2 v 2.4 [ P = .0002]; CNV, 42.5 v 2.0 [ P = .0003]). GCN correlated with CNV in both cohorts (exploratory, r = 0.90; validation, r = 0.97; P < .0001). CNV in cross-validation of the 2 NGS panels also showed a strong correlation ( r = 0.98; P < .0001). CNV in patients who fulfilled the consensus criteria was > 4.0 in all, which demonstrates the accuracy of the IHC/FISH criteria and cross-validation of NGS panels. CONCLUSION We have established and verified harmonized diagnostic criteria for HER2+ and demonstrated consistency between IHC/FISH and CNV determined by NGS in mCRC.
Collapse
Affiliation(s)
- Satoshi Fujii
- Division of Pathology, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, Kashiwa, Japan
| | | | - Jihun Kim
- Department of Pathology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, South Korea
| | - Wataru Okamoto
- Biobank Translational Research Support Section, Translational Research Management Division, Clinical Research Support Office, National Cancer Center Hospital East, Kashiwa, Japan
| | - Jeong Eun Kim
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Kentaro Sawada
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Yoshiaki Nakamura
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Scott Kopetz
- Department of Gastrointestinal Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Woong-Yang Park
- Samsung Genome Institute, Samsung Medical Center, Seoul, South Korea
| | - Katsuya Tsuchihara
- Division of Translational Informatics, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, Kashiwa, Japan
| | - Tae Won Kim
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Kanwal Raghav
- Department of Gastrointestinal Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Takayuki Yoshino
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| |
Collapse
|
214
|
Zhang W, Zhou H, Wang Y, Zhang Z, Cao G, Song T, Zhang T, Li Q. Systemic treatment of advanced or recurrent biliary tract cancer. Biosci Trends 2020; 14:328-341. [PMID: 32830166 DOI: 10.5582/bst.2020.03240] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Affiliation(s)
- Wei Zhang
- Department of Hepatobiliary Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin. Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Hongyuan Zhou
- Department of Hepatobiliary Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin. Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Yingying Wang
- Department of Hepatobiliary Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin. Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Zewu Zhang
- Department of Hepatobiliary Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin. Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Guangtai Cao
- Cangzhou Hospital of Integrated TCM-WM Cangzhou, Hebei, China
| | - Tianqiang Song
- Department of Hepatobiliary Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin. Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Ti Zhang
- Department of Hepatobiliary Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin. Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Qiang Li
- Department of Hepatobiliary Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin. Tianjin's Clinical Research Center for Cancer, Tianjin, China
| |
Collapse
|
215
|
Zhang Q, Fu Q, Bai X, Liang T. Molecular Profiling-Based Precision Medicine in Cancer: A Review of Current Evidence and Challenges. Front Oncol 2020; 10:532403. [PMID: 33194591 PMCID: PMC7652987 DOI: 10.3389/fonc.2020.532403] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Accepted: 08/26/2020] [Indexed: 12/16/2022] Open
Abstract
Matched therapy based on next-generation sequencing is now a part of routine care to guide the treatment of patients with advanced solid tumors. However, whether and to what extent patients can benefit from this strategy on a large scale remains uncertain. In the past decade, several clinical studies were performed in this field, among which only one was a randomized trial. We reviewed the literature on this topic and summarize the existing data about the efficacy of this treatment strategy. Currently, the evidence is promising but not solid. Multiple ongoing trials are also summarized. We also discuss the limitations of this treatment strategy and certain unsolved important problems, including how to select the sample and target level, how to interpret the results, and the problem of drug accessibility. All these issues should receive more attention in future clinical trial design and the application of target therapy in cancer treatment.
Collapse
Affiliation(s)
- Qi Zhang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- The Key Laboratory of Pancreatic Diseases of Zhejiang Province, Hangzhou, China
- The Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou, China
- Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Hangzhou, China
| | - Qihan Fu
- The Key Laboratory of Pancreatic Diseases of Zhejiang Province, Hangzhou, China
- The Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou, China
- Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Hangzhou, China
- Department of Medical Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xueli Bai
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- The Key Laboratory of Pancreatic Diseases of Zhejiang Province, Hangzhou, China
- The Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou, China
- Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Hangzhou, China
| | - Tingbo Liang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- The Key Laboratory of Pancreatic Diseases of Zhejiang Province, Hangzhou, China
- The Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou, China
- Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Hangzhou, China
| |
Collapse
|
216
|
Salem ME, Puccini A, Tie J. Redefining Colorectal Cancer by Tumor Biology. Am Soc Clin Oncol Educ Book 2020; 40:1-13. [PMID: 32207671 DOI: 10.1200/edbk_279867] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Colorectal cancer treatment has undergone a paradigm shift. We no longer see this disease as a singular, anatomic tumor type but rather a set of disease subgroups. Largely because of a better understanding of cancer biology and the introduction and integration of molecular biomarkers-the premise of precision therapy-we are beginning to direct treatments toward the right tumor target(s) in the right patients. The field of molecular profiling is continually evolving, and new biomarkers are constantly being discovered that have investigational, therapeutic, and/or prognostic implications-negative or positive. To date, only a few biomarkers have sufficient actionable, clinical implication to earn international guideline-recommended routine testing. Hence, it is vital that the treating oncologist should know which biomarkers to assess, when in the treatment course to test for them, and how the test is to be done. Correct interpretation of profiling results is imperative. Herein, we focus on international guideline-recommended mutation testing for patients prior to their colorectal cancer treatment initiation. The clinical applications of circulating tumor DNA (ctDNA) in patients with metastatic disease, based on our current knowledge and capabilities, are also addressed.
Collapse
Affiliation(s)
- Mohamed E Salem
- Department of Medical Oncology, Levine Cancer Institute, Charlotte, NC
| | - Alberto Puccini
- University of Genoa, Ospedale Policlinico San Martino IRCCS, Genoa, Italy
| | - Jeanne Tie
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.,Division of Personalized Oncology, The Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia
| |
Collapse
|
217
|
Di Villeneuve L, Souza IL, Tolentino FDS, Ferrarotto R, Schvartsman G. Salivary Gland Carcinoma: Novel Targets to Overcome Treatment Resistance in Advanced Disease. Front Oncol 2020; 10:580141. [PMID: 33194707 PMCID: PMC7649804 DOI: 10.3389/fonc.2020.580141] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Accepted: 09/09/2020] [Indexed: 12/20/2022] Open
Abstract
Salivary gland carcinomas (SGCs) account for <5% of head and neck malignant neoplasms, further subcategorized in over 20 histological subtypes. For the most part, treatment for advanced disease is guided by morphology. SGCs in general respond poorly to a wide array of standard chemotherapy, with short durability, and significant toxicity. More recently, next-generation sequencing provided significant input on the molecular characterization of each SGC subtype, not only improving diagnostic differentiation between morphologically similar tumor types but also identifying novel driver pathways that determine tumor biology and may be amenable to targeted therapy. Among the most common histological subtype is adenoid cystic carcinoma, which often harbors a chromosome translocation resulting in an MYB-NFIB oncogene, with various degrees of Myb surface expression. In a smaller subset, NOTCH1 mutations occur, conferring a more aggressive pattern and potential sensitivity to Notch inhibitors. Salivary duct carcinomas may overexpress Her-2 and androgen receptors, with promising clinical outcomes after exposure to targeted therapies approved for other indications. Secretory carcinoma, previously known as mammary analog secretory carcinoma, is distinguished by an ETV6-NTRK3 fusion that can both help differentiate it from its morphologically similar acinar cell carcinoma and make it susceptible to Trk inhibitors. In the present article, we discuss the molecular abnormalities, their impact on tumor biology, and therapeutic opportunities for the most common SGC subtypes and review published and ongoing clinical trials and future perspectives for this rare disease.
Collapse
Affiliation(s)
- Larissa Di Villeneuve
- Department of Medical Oncology, Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - Ive Lima Souza
- Department of Medical Oncology, Hospital Israelita Albert Einstein, São Paulo, Brazil
| | | | - Renata Ferrarotto
- Department of Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Gustavo Schvartsman
- Department of Medical Oncology, Hospital Israelita Albert Einstein, São Paulo, Brazil
| |
Collapse
|
218
|
Sasankan S, Rebuck L, Darrah G, Harari Turquie M, Rabinowitz I. Metastatic Pancreatic Cancer with BRAF and P53 Mutations: Case Report of Therapeutic Response to Doublet Targeted Therapy. Case Rep Oncol 2020; 13:1239-1243. [PMID: 33250737 PMCID: PMC7670384 DOI: 10.1159/000510096] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Accepted: 07/13/2020] [Indexed: 12/23/2022] Open
Abstract
We report on the clinical history of a 49-year-old female with metastatic pancreatic cancer. She was initially treated with standard chemotherapy as per current guidelines. She was found to have both a BRAF and P53 mutation, and received dabrafenib and trametinib with deep responses, both radiographically and biochemically (CA19-9). Her response has been more clinically relevant than responses in previous case reports of patients with BRAF-positive pancreatic cancer treated with targeted therapy. To the best of our knowledge, this is the first case report showing a dramatic therapeutic response to combination therapy with dabrafenib and trametinib in metastatic pancreatic cancer.
Collapse
Affiliation(s)
- Shenthol Sasankan
- Hematology and Medical Oncology, University of New Mexico Comprehensive Cancer Center, Albuquerque, New Mexico, USA
| | - Lorraine Rebuck
- Hematology and Medical Oncology, University of New Mexico Comprehensive Cancer Center, Albuquerque, New Mexico, USA
| | - Gloria Darrah
- Hematology and Medical Oncology, University of New Mexico Comprehensive Cancer Center, Albuquerque, New Mexico, USA
| | - Moises Harari Turquie
- Hematology and Medical Oncology, University of New Mexico Comprehensive Cancer Center, Albuquerque, New Mexico, USA
| | - Ian Rabinowitz
- Hematology and Medical Oncology, University of New Mexico Comprehensive Cancer Center, Albuquerque, New Mexico, USA
| |
Collapse
|
219
|
Flaherty KT, Gray RJ, Chen AP, Li S, McShane LM, Patton D, Hamilton SR, Williams PM, Iafrate AJ, Sklar J, Mitchell EP, Harris LN, Takebe N, Sims DJ, Coffey B, Fu T, Routbort M, Zwiebel JA, Rubinstein LV, Little RF, Arteaga CL, Comis R, Abrams JS, O'Dwyer PJ, Conley BA. Molecular Landscape and Actionable Alterations in a Genomically Guided Cancer Clinical Trial: National Cancer Institute Molecular Analysis for Therapy Choice (NCI-MATCH). J Clin Oncol 2020; 38:3883-3894. [PMID: 33048619 PMCID: PMC7676882 DOI: 10.1200/jco.19.03010] [Citation(s) in RCA: 195] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Therapeutically actionable molecular alterations are widely distributed across cancer types. The National Cancer Institute Molecular Analysis for Therapy Choice (NCI-MATCH) trial was designed to evaluate targeted therapy antitumor activity in underexplored cancer types. Tumor biopsy specimens were analyzed centrally with next-generation sequencing (NGS) in a master screening protocol. Patients with a tumor molecular alteration addressed by a targeted treatment lacking established efficacy in that tumor type were assigned to 1 of 30 treatments in parallel, single-arm, phase II subprotocols.
Collapse
Affiliation(s)
| | - Robert J Gray
- ECOG-ACRIN Cancer Research Group Biostatistics Center, Dana Farber Cancer Institute Boston, MA
| | - Alice P Chen
- Division of Cancer Treatment and Diagnosis, National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD
| | - Shuli Li
- ECOG-ACRIN Cancer Research Group Biostatistics Center, Dana Farber Cancer Institute Boston, MA
| | - Lisa M McShane
- Division of Cancer Treatment and Diagnosis, National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD
| | - David Patton
- Center for Biomedical Informatics and Information Technology, NCI, NIH, Bethesda, MD
| | | | | | - A John Iafrate
- Massachusetts General Hospital, Boston, MA.,Harvard University, Boston, MA
| | | | | | - Lyndsay N Harris
- Division of Cancer Treatment and Diagnosis, National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD
| | - Naoko Takebe
- Division of Cancer Treatment and Diagnosis, National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD
| | - David J Sims
- Frederick National Laboratory for Cancer Research, Frederick, MD
| | - Brent Coffey
- Center for Biomedical Informatics and Information Technology, Frederick National Laboratory for Cancer Research, Frederick, MD
| | - Tony Fu
- Frederick National Laboratory for Cancer Research, Frederick, MD
| | - Mark Routbort
- University of Texas MD Anderson Cancer Center, Houston, TX
| | - James A Zwiebel
- Division of Cancer Treatment and Diagnosis, National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD
| | - Larry V Rubinstein
- Division of Cancer Treatment and Diagnosis, National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD
| | - Richard F Little
- Division of Cancer Treatment and Diagnosis, National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD
| | - Carlos L Arteaga
- University of Texas Southwestern Simmons Cancer Center, Dallas, TX
| | - Robert Comis
- ECOG-ACRIN Cancer Research Group, Philadelphia, PA.,Deceased
| | - Jeffrey S Abrams
- Division of Cancer Treatment and Diagnosis, National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD
| | - Peter J O'Dwyer
- Division of Cancer Treatment and Diagnosis, National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD
| | - Barbara A Conley
- Division of Cancer Treatment and Diagnosis, National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD
| | | |
Collapse
|
220
|
Yang ZY, Huang JH, Chen B, Xu CW, Lei L, Wang XJ, Fang MY. Efficacy of Pyrotinib in HER2-Overexpressing Salivary Duct Carcinoma With Lung Metastasis: A Case Report. Front Oncol 2020; 10:559057. [PMID: 33123470 PMCID: PMC7567027 DOI: 10.3389/fonc.2020.559057] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 08/27/2020] [Indexed: 12/27/2022] Open
Abstract
Background: Salivary duct carcinoma (SDC), an aggressive and rare malignancy with poor prognosis, is mostly associated with the overexpression of the androgen receptor (AR) and human epidermal growth factor receptor 2 (HER2). However, limited data are available for the targeting of both HER2 and AR in advanced/metastatic SDC. Case Presentation: A 62-year-old man with advanced SDC accompanied by lung and lymph node metastasis showed disease progression after two lines of chemotherapy and endocrine therapy. Metastatic lesions from the lung biopsy were obtained, and immunohistochemistry (IHC) indicated the overexpression of AR and HER2 (3+). The patient was administered pyrotinib (a pan-ErbB receptor tyrosine kinase inhibitor) and bicalutamide (an androgen receptor antagonist) as a third-line treatment. During the ten months of follow-up, a durable partial response was achieved with this combination. Conclusions: This is the first clinical study to report the successful application of pyrotinib in a patient with advanced SDC. We recommend that pyrotinib and bicalutamide be used as salvage therapy for AR and HER2-positive advanced metastases in SDC, given the favorable response and clinical benefit.
Collapse
Affiliation(s)
- Zi-Yan Yang
- Department of Breast Oncology, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, China.,Institute of Cancer and Basic Medicine (ICBM), Chinese Academy of Sciences, Hangzhou, China.,Department of Oncology, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, China
| | - Jia-Huan Huang
- Department of Breast Oncology, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, China.,Institute of Cancer and Basic Medicine (ICBM), Chinese Academy of Sciences, Hangzhou, China
| | - Bo Chen
- Department of Pathology, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, China
| | - Chun-Wei Xu
- Department of Respiratory, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Lei Lei
- Department of Breast Oncology, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, China.,Institute of Cancer and Basic Medicine (ICBM), Chinese Academy of Sciences, Hangzhou, China
| | - Xiao-Jia Wang
- Department of Breast Oncology, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, China.,Institute of Cancer and Basic Medicine (ICBM), Chinese Academy of Sciences, Hangzhou, China
| | - Mei-Yu Fang
- Department of General Oncology, Key Laboratory of Head and Neck Cancer Translational Research of Zhejiang Province, Institute of Cancer Research and Basic Medical Sciences of Chinese Academy of Sciences, Cancer Hospital of the University of Chinese Academy of Sciences, Hangzhou, China
| |
Collapse
|
221
|
Nakaguro M, Tada Y, Faquin WC, Sadow PM, Wirth LJ, Nagao T. Salivary duct carcinoma: Updates in histology, cytology, molecular biology, and treatment. Cancer Cytopathol 2020; 128:693-703. [PMID: 32421944 PMCID: PMC7541685 DOI: 10.1002/cncy.22288] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 03/31/2020] [Accepted: 04/21/2020] [Indexed: 12/15/2022]
Abstract
Salivary duct carcinoma (SDC) is an aggressive subtype of primary salivary gland carcinoma, often with an advanced stage at presentation and high rates of metastasis and recurrence. It most commonly arises in the parotid gland of older men and microscopically resembles high-grade breast ductal carcinoma. While 50 years have lapsed since the first report of this entity, recent intensive studies have shed light on its biologic, genetic, and clinical characteristics. The diagnosis of SDC is aided by the immunohistochemical expression of androgen receptor (AR) coupled with its characteristic histomorphology. Fine-needle aspiration typically reveals cytologic features of high-grade carcinoma, and ancillary studies using cell block material can facilitate the specific diagnosis of SDC. In surgical specimens, certain histologic features are important prognostic factors, including nuclear pleomorphism, mitotic counts, vascular invasion, and the morphology at the invasion front. Several clinical studies have shown promising results using targeted therapy for AR and human epidermal growth factor receptor 2 (HER2), and the latest version of the National Comprehensive Cancer Network guidelines recommends the evaluation of AR and HER2 status before treatment. Recent molecular analyses have revealed multiple heterogeneous alterations in well-known oncogenes and tumor suppressor genes, including TP53, HRAS, PIK3CA, PTEN, and BRAF. Clinical trials of drugs targeting these genes may broaden the treatment options for SDC in the near future.
Collapse
Affiliation(s)
- Masato Nakaguro
- Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
- Department of Pathology and Laboratory Medicine, Nagoya University Hospital, Nagoya, Japan
| | - Yuichiro Tada
- Department of Head and Neck Oncology and Surgery, International University of Health and Welfare Mita Hospital, Tokyo, Japan
| | - William C. Faquin
- Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Peter M. Sadow
- Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Lori J. Wirth
- Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Toshitaka Nagao
- Department of Anatomic Pathology, Tokyo Medical University, Tokyo, Japan
| |
Collapse
|
222
|
Li A, Bergan RC. Clinical trial design: Past, present, and future in the context of big data and precision medicine. Cancer 2020; 126:4838-4846. [PMID: 32931022 PMCID: PMC7693060 DOI: 10.1002/cncr.33205] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 08/17/2020] [Accepted: 08/20/2020] [Indexed: 12/15/2022]
Abstract
Clinical trials are fundamental for advances in cancer treatment. The traditional framework of phase 1 to 3 trials is designed for incremental advances between regimens. However, our ability to understand and treat cancer has evolved with the increase in drugs targeting an expanding array of therapeutic targets, the development of progressively comprehensive data sets, and emerging computational analytics, all of which are reshaping our treatment strategies. A more robust linkage between drugs and underlying cancer biology is blurring historical lines that define trials on the basis of cancer type. The complexity of the molecular basis of cancer, coupled with manifold variations in clinical status, is driving the individually tailored use of combinations of precision targeted drugs. This approach is spawning a new era of clinical trial types. Although most care is delivered in a community setting, large centers support real‐time multi‐omic analytics and their integrated interpretation by using machine learning in the context of real‐world data sets. Coupling the analytic capabilities of large centers to the tailored delivery of therapy in the community is forging a paradigm that is optimizing service for patients. Understanding the importance of these evolving trends across the health care spectrum will affect our treatment of cancer in the future and is the focus of this review. With advances in cancer biology, precision therapeutics, and big data, clinical trial designs are evolving. They are transforming cancer care and research across the biomedical enterprise.
Collapse
Affiliation(s)
- Allen Li
- Division of Hematology/Oncology, Knight Cancer Institute, Oregon Health and Science University, Portland, Oregon
| | - Raymond C Bergan
- Division of Hematology/Oncology, Knight Cancer Institute, Oregon Health and Science University, Portland, Oregon
| |
Collapse
|
223
|
Hiraoka N, Nitta H, Ohba A, Yoshida H, Morizane C, Okusaka T, Nara S, Esaki M, Kishi Y, Shimada K. Details of human epidermal growth factor receptor 2 status in 454 cases of biliary tract cancer. Hum Pathol 2020; 105:9-19. [PMID: 32891647 DOI: 10.1016/j.humpath.2020.08.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 08/25/2020] [Accepted: 08/25/2020] [Indexed: 12/20/2022]
Abstract
Human epidermal growth factor receptor 2 (HER2)-targeted therapy has improved clinical outcomes in patients with HER2-positive breast and gastric cancers, although ineffective or recurrent cases are present. One reason for this is the heterogeneity of HER2 expression in cancer cells. The aim of this study was to investigate the clinicopathological characteristics and HER2 status of patients with biliary tract cancers (BTCs). We examined HER2 protein expression by immunohistochemistry, HER2 gene amplification by fluorescence in situ hybridization, and both HER2 protein and gene levels simultaneously by gene-protein assay. Samples were collected from 454 patients who underwent surgical resection for BTCs (110 intrahepatic cholangiocarcinomas [ICC], 67 perihilar extrahepatic cholangiocarcinomas [ECC-Bp], 119 distal extrahepatic cholangiocarcinomas [ECC-Bd], 80 gallbladder carcinomas [GBC], and 79 ampullary carcinomas [AVC]). HER2 status was assessed according to the guidelines for HER2 testing in gastroesophageal adenocarcinoma. HER2-positive status was detected in 14.5% of BTCs (3.7% of ICC, 3.0% of ECC-Bp, 18.5% of ECC-Bd, 31.3% of GBC, and 16.4% of AVC). Furthermore, HER2-positivity tended to correlate with low histological grade, tumor histology, and macroscopic features in certain tumors. HER2 heterogeneity was common and highly frequent (83%) in BTC cases. Reduced HER2 protein expression in the deeper invasive areas with simultaneous dedifferentiation was frequently observed in HER2-positive cancer cells. The findings of this study suggest that a large subgroup of HER2-positive BTC cases can be considered for HER2-targeted therapy. Moreover, the HER2 status in BTCs should be determined carefully using a sensitive approach toward larger cancer tissues.
Collapse
Affiliation(s)
- Nobuyoshi Hiraoka
- Division of Pathology and Clinical Laboratories, National Cancer Center Hospital, Tokyo, 104-0045, Japan.
| | | | - Akihiro Ohba
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital, Tokyo, 104-0045, Japan
| | - Hiroshi Yoshida
- Division of Pathology and Clinical Laboratories, National Cancer Center Hospital, Tokyo, 104-0045, Japan
| | - Chigusa Morizane
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital, Tokyo, 104-0045, Japan
| | - Takuji Okusaka
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital, Tokyo, 104-0045, Japan
| | - Satoshi Nara
- Hepatobiliary and Pancreatic Surgery Division, National Cancer Center Hospital, Tokyo, 104-0045, Japan
| | - Minoru Esaki
- Hepatobiliary and Pancreatic Surgery Division, National Cancer Center Hospital, Tokyo, 104-0045, Japan
| | - Yoji Kishi
- Hepatobiliary and Pancreatic Surgery Division, National Cancer Center Hospital, Tokyo, 104-0045, Japan
| | - Kazuaki Shimada
- Hepatobiliary and Pancreatic Surgery Division, National Cancer Center Hospital, Tokyo, 104-0045, Japan
| |
Collapse
|
224
|
Eskander RN, Elvin J, Gay L, Ross JS, Miller VA, Kurzrock R. Unique Genomic Landscape of High-Grade Neuroendocrine Cervical Carcinoma: Implications for Rethinking Current Treatment Paradigms. JCO Precis Oncol 2020; 4:1900248. [PMID: 33015532 PMCID: PMC7529537 DOI: 10.1200/po.19.00248] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/09/2020] [Indexed: 12/18/2022] Open
Abstract
PURPOSE High-grade neuroendocrine cervical cancer (HGNECC) is an uncommon malignancy with limited therapeutic options; treatment is patterned after the histologically similar small-cell lung cancer (SCLC). To better understand HGNECC biology, we report its genomic landscape. PATIENTS AND METHODS Ninety-seven patients with HGNECC underwent comprehensive genomic profiling (182-315 genes). These results were subsequently compared with a cohort of 1,800 SCLCs. RESULTS The median age of patients with HGNECC was 40.5 years; 83 patients (85.6%) harbored high-risk human papillomavirus (HPV). Overall, 294 genomic alterations (GAs) were identified (median, 2 GAs/sample; average, 3.0 GAs/sample, range, 0-25 GAs/sample) in 109 distinct genes. The most frequently altered genes were PIK3CA (19.6% of cohort), MYC (15.5%), TP53 (15.5%), and PTEN (14.4%). RB1 GAs occurred in 4% versus 32% of HPV-positive versus HPV-negative tumors (P < .0001). GAs in HGNECC involved the following pathways: PI3K/AKT/mTOR (41.2%); RAS/MEK (11.3%); homologous recombination (9.3%); and ERBB (7.2%). Two tumors (2.1%) had high tumor mutational burden (TMB; both with MSH2 alterations); 16 (16.5%) had intermediate TMB. Seventy-one patients (73%) had ≥ 1 alteration that was theoretically druggable. Comparing HGNECC with SCLC, significant differences in TMB, microsatellite instability, HPV-positive status, and in PIK3CA, MYC, PTEN, TP53, ARID1A, and RB1 alteration rates were found. CONCLUSION This large cohort of patients with HGNECC demonstrated a genomic landscape distinct from SCLC, calling into question the biologic and therapeutic relevance of the histologic similarities between the entities. Furthermore, 73% of HGNECC tumors had potentially actionable alterations, suggesting novel treatment strategies for this aggressive malignancy.
Collapse
Affiliation(s)
- Ramez N Eskander
- Department of Obstetrics, Gynecology and Reproductive Sciences, Division of Gynecologic Oncology, University of California San Diego, La Jolla, CA.,Center for Personalized Cancer Therapy, University of California San Diego, La Jolla, CA
| | | | | | - Jeffrey S Ross
- Foundation Medicine, Cambridge, MA.,Upstate Medical University, Syracuse, NY
| | | | - Razelle Kurzrock
- Center for Personalized Cancer Therapy, University of California San Diego, La Jolla, CA.,Department of Medicine, Division of Hematology/Oncology, University of California San Diego, La Jolla, CA
| |
Collapse
|
225
|
Fernández-Montes A, Grávalos C, Pericay C, Safont MJ, Benavides M, Élez E, García-Alfonso P, García-Paredes B, Carrato A, Aranda E. Current Options for Third-line and Beyond Treatment of Metastatic Colorectal Cancer. Spanish TTD Group Expert Opinion. Clin Colorectal Cancer 2020; 19:165-177. [PMID: 32507561 DOI: 10.1016/j.clcc.2020.04.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 04/08/2020] [Accepted: 04/13/2020] [Indexed: 12/13/2022]
Abstract
Colorectal cancer (CRC) is a public health problem: it is the third most common cancer in men (746,000 new cases/year) and the second in women (614,000 new cases/year), representing the second leading cause of death by cancer worldwide. The survival of patients with metastatic CRC (mCRC) has increased prominently in recent years, reaching a median of 25 to 30 months. A growing number of patients with mCRC are candidates to receive a treatment in third line or beyond, although the optimal drug regimen and sequence are still unknown. In this situation of refractoriness, there are several alternatives: (1) To administer sequentially the 2 oral drugs approved in this indication: trifluridine/tipiracil and regorafenib, which have shown a statistically significant benefit in progression-free survival and overall survival with a different toxicity profile. (2) To administer cetuximab or panitumumab in treatment-naive patients with RAS wild type, which is increasingly rare because these drugs are usually indicated in first- or second-line. (3) To reuse drugs already administered that were discontinued owing to toxicity or progression (oxaliplatin, irinotecan, fluoropyrimidine, antiangiogenics, anti-epidermal growth factor receptor [if RAS wild-type]). High-quality evidence is limited, but this strategy is often used in routine clinical practice in the absence of alternative therapies especially in patients with good performance status. (4) To use specific treatments for very selected populations, such as trastuzumab/lapatinib in mCRC human epidermal growth factor receptor 2-positive, immunotherapy in microsatellite instability, intrahepatic therapies in limited disease or primarily located in the liver, although the main recommendation is to include patients in clinical trials.
Collapse
Affiliation(s)
- Ana Fernández-Montes
- Medical Oncology, Complejo Hospitalario Universitario de Ourense, Orense, Spain.
| | - Cristina Grávalos
- Medical Oncology, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Carles Pericay
- Medical Oncology, Hospital de Sabadell, Corporación Sanitaria Parc Tauli, Sabadell, Barcelona, Spain
| | - Ma José Safont
- Medical Oncology, Hospital General Universitario de Valencia, València, Spain
| | - Manuel Benavides
- Medical Oncology, Hospital Universitario Regional y Virgen de la Victoria, Málaga, Spain
| | - Elena Élez
- Medical Oncology, Hospital Universitari Vall d'Hebron, Passeig de la Vall d'Hebron, 119-129, 08035 Barcelona, Spain
| | | | - Beatriz García-Paredes
- Medical Oncology, Hospital Clínico San Carlos, Instituto de Investigación Hospital Clínico San Carlos (IdISSC). CIBERONC, Madrid, Spain
| | - Alfredo Carrato
- Medical Oncology, Hospital Universitario Ramón y Cajal, Universidad de Alcalá de Henares, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), CIBERONC, Madrid, Spain
| | - Enrique Aranda
- Medical Oncology, Hospital Reina Sofía, University of Córdoba, Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), CIBERONC, Córdoba, Spain
| |
Collapse
|
226
|
Saotome K, Chiyoda T, Aimono E, Nakamura K, Tanishima S, Nohara S, Okada C, Hayashi H, Kuroda Y, Nomura H, Susumu N, Iwata T, Yamagami W, Kataoka F, Nishihara H, Aoki D. Clinical implications of next-generation sequencing-based panel tests for malignant ovarian tumors. Cancer Med 2020; 9:7407-7417. [PMID: 32813918 PMCID: PMC7571820 DOI: 10.1002/cam4.3383] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 07/23/2020] [Accepted: 07/25/2020] [Indexed: 12/29/2022] Open
Abstract
Precision medicine based on cancer genomics is being applied in clinical practice. However, patients do not always derive benefits from genomic testing. Here, we performed targeted amplicon exome sequencing‐based panel tests, including 160 cancer‐related genes (PleSSision‐160), on 88 malignant ovarian tumors (high‐grade serous carcinoma, 27; endometrioid carcinoma, 15; clear cell carcinoma, 30; mucinous carcinoma, 6; undifferentiated carcinoma, 4; and others, 6 (immature teratoma, 1; carcinosarcoma, 3; squamous cell carcinoma, 1; and mixed, 1)), to assess treatment strategies and useful biomarkers for malignant ovarian tumors. Overall, actionable gene variants were found in 90.9%, and druggable gene variants were found in 40.9% of the cases. Actionable BRCA1 and BRCA2 variants were found in 4.5% of each of the cases. ERBB2 amplification was found in 33.3% of mucinous carcinoma cases. Druggable hypermutation/ultramutation (tumor mutation burden ≥ 10 SNVs/Mbp) was found in 7.4% of high‐grade serous carcinoma, 46.7% of endometrioid carcinoma, 10% of clear cell carcinoma, 0% of mucinous carcinoma, 25% of undifferentiated carcinoma, and 33.3% of the other cancer cases. Copy number alterations were significantly higher in high‐grade serous carcinoma (P < .005) than in other histologic subtypes; some clear cell carcinoma showed high copy number alterations that were correlated with advanced stage (P < .05) and worse survival (P < .01). A high count of copy number alteration was associated with worse survival in all malignant ovarian tumors (P < .05). Our study shows that targeted agents can be detected in approximately 40% of malignant ovarian tumors via multigene panel testing, and copy number alteration count can be a useful marker to help assess risks in malignant ovarian tumor patients.
Collapse
Affiliation(s)
- Keiko Saotome
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo, Japan
| | - Tatsuyuki Chiyoda
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo, Japan
| | - Eriko Aimono
- Genomics Unit, Keio Cancer Center, Keio University School of Medicine, Tokyo, Japan
| | - Kohei Nakamura
- Genomics Unit, Keio Cancer Center, Keio University School of Medicine, Tokyo, Japan
| | - Shigeki Tanishima
- Department of Biomedical Informatics Development, Mitsubishi Space Software Co., Ltd, Amagasaki, Japan
| | - Sachio Nohara
- Department of Biomedical Informatics Development, Mitsubishi Space Software Co., Ltd, Amagasaki, Japan
| | - Chihiro Okada
- Department of Biomedical Informatics Development, Mitsubishi Space Software Co., Ltd, Amagasaki, Japan
| | - Hideyuki Hayashi
- Genomics Unit, Keio Cancer Center, Keio University School of Medicine, Tokyo, Japan
| | - Yuka Kuroda
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo, Japan
| | - Hiroyuki Nomura
- Department of Obstetrics and Gynecology, School of Medicine, Fujita Health University, Toyoake, Japan
| | - Nobuyuki Susumu
- Department of Obstetrics and Gynecology, International University of Health and Welfare, School of Medicine, Narita, Japan
| | - Takashi Iwata
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo, Japan
| | - Wataru Yamagami
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo, Japan
| | - Fumio Kataoka
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo, Japan
| | - Hiroshi Nishihara
- Genomics Unit, Keio Cancer Center, Keio University School of Medicine, Tokyo, Japan
| | - Daisuke Aoki
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
227
|
Ryan ÉJ, Creavin B, Sheahan K. Delivery of Personalized Care for Locally Advanced Rectal Cancer: Incorporating Pathological, Molecular Genetic, and Immunological Biomarkers Into the Multimodal Paradigm. Front Oncol 2020; 10:1369. [PMID: 32923389 PMCID: PMC7456909 DOI: 10.3389/fonc.2020.01369] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 06/29/2020] [Indexed: 12/13/2022] Open
Abstract
Approximately one-third of all newly diagnosed colorectal cancer (CRC) is composed of rectal cancer, with the incidence rising in younger patients. The principal neoadjuvant treatments consist of neoadjuvant short-course radiotherapy and long-course chemoradiation. Locally advanced rectal cancer (LARC) is particularly challenging to manage given the anatomical constrictions of the pelvis and the risk for local recurrence. In appropriately treated patients, 5- and 10-year overall survival is estimated at 60 and 50%, respectively. The prognosis for LARC has improved in recent years with more access to screening, advances in surgical techniques, and perioperative care. Furthermore, the refinement of the multidisciplinary team with combined-modality management strategies has improved outcomes. These advancements have been augmented by significant improvements in the understanding of the underlying tumor biology. However, there are many instances where patient outcomes do not match those for their tumor stage and accurate prognostic information for individual patients can be difficult to estimate owing to the heterogeneous nature of LARC. Many new combinations of chemotherapy with radiotherapy, including total neoadjuvant therapy with targeted therapies that aim to diminish toxicity and increase survival, are being evaluated in clinical trials. Despite these advances, local recurrence and distant metastasis remain an issue, with one-third of LARC patients dying within 5 years of initial treatment. Although much of the new pathological, molecular genetics, and immunological biomarkers allow refinement in the classification and prognostication of CRC, the relative importance of each of these factors with regards to the development and progression of LARC remains incompletely understood. These factors are often insufficiently validated and seldom consider the individual characteristics of the host, the tumor and its location, the local available expertise, or the probable location of recurrence. Appreciating the mechanisms behind these differences will allow for a more comprehensive, personalized approach and more informed treatment options, leading to ultimately superior outcomes. This review aims to first outline the current multidisciplinary context in which LARC care should be delivered and then discuss how some key prognosticators, including novel histopathological, molecular genetics, and immunological biomarkers, might fit into the wider context of personalized LARC management in the coming years.
Collapse
Affiliation(s)
- Éanna J. Ryan
- School of Medicine, University College Dublin, Dublin, Ireland
- Department of Surgery, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Ben Creavin
- School of Medicine, University College Dublin, Dublin, Ireland
- Department of Surgery, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Kieran Sheahan
- School of Medicine, University College Dublin, Dublin, Ireland
| |
Collapse
|
228
|
Takahashi K, Ishibashi E, Kubo T, Harada Y, Hayashi H, Kano M, Shimizu Y, Shirota H, Mori Y, Muto M, Ishioka C, Dosaka-Akita H, Matsubara H, Nishihara H, Sueoka-Aragane N, Toyooka S, Hirakawa A, Tateishi U, Miyake S, Ikeda S. A phase 2 basket trial of combination therapy with trastuzumab and pertuzumab in patients with solid cancers harboring human epidermal growth factor receptor 2 amplification (JUPITER trial). Medicine (Baltimore) 2020; 99:e21457. [PMID: 32769873 PMCID: PMC7592999 DOI: 10.1097/md.0000000000021457] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
INTRODUCTION Human epidermal growth factor receptor 2 (HER2) gene amplification and mutations have emerged as oncogenic drivers and therapeutic targets not limited to breast and gastric cancers, but also in a variety of cancers. However, even if an actionable gene alteration is found, the incidence of HER2 amplification in these cancers is less than 5%. It is too difficult to conduct a conventional randomized, controlled trial in a rare fraction. Therefore, we have designed a organ-agnostic basket study, which covers a variety of solid cancers harboring HER2 amplification, in 1 study protocol. METHODS/DESIGN This trial is a multicenter, single-arm, basket phase 2 study in Japan. Patients with solid cancers harboring HER2 amplification that have progressed with standard treatment, or rare cancers for which there is no standard treatment, will be eligible. Target cancers include bile duct, urothelial, uterine, ovarian, and other solid cancers where HER2 amplification is detected by comprehensive genomic profiling using next-generation sequencing technology. A total of 38 patients will be treated with combination therapy with trastuzumab and pertuzumab every 3 weeks until disease progression, unmanageable toxicity, death, or patient refusal. The primary endpoint is the objective response rate, and secondary endpoints are progression-free survival, overall survival, and duration of response. DISCUSSION The aim of this trial is to evaluate the safety and efficacy of combination therapy with trastuzumab and pertuzumab in patients with locally advanced or metastatic, solid cancers harboring HER2 amplification. Instead of focusing on 1 organ type, our trial design uses a basket study focusing on HER2 amplification, regardless of the site or origin of the cancer. The results of our study will advance clinical and scientific knowledge concerning the treatment of locally advanced, rare solid cancers harboring HER2 amplification, using the combination of trastuzumab and pertuzumab. TRIAL REGISTRATION This trial was registered in Japan Registry of Clinical Trials (jCRT) on February 25, 2019, as jRCT2031180150.
Collapse
Affiliation(s)
| | - Eri Ishibashi
- Medical Innovation Promotion Center, Tokyo Medical and Dental University, Tokyo
| | - Toshio Kubo
- Department of General Thoracic Surgery and Breast and Endocrinological Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Science, Okayama
| | - Yohei Harada
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Faculty of Medicine, Saga University, Saga
| | - Hideyuki Hayashi
- Genomics Unit, Keio Cancer Center, Keio University School of Medicine, Tokyo
| | - Masayuki Kano
- Department of Frontier Surgery, Chiba University, Chiba
| | - Yasushi Shimizu
- Department of Medical Oncology, Faculty of Medicine & Graduate School of Medicine, Hokkaido University, Hokkaido
| | - Hidekazu Shirota
- Department of Clinical Oncology, Tohoku University Hospital, Sendai
| | - Yukiko Mori
- Department of Therapeutic Oncology, Graduate School of Medicine, Kyoto University, Kyoto
| | - Manabu Muto
- Department of Therapeutic Oncology, Graduate School of Medicine, Kyoto University, Kyoto
| | - Chikashi Ishioka
- Department of Clinical Oncology, Tohoku University Hospital, Sendai
| | - Hirotoshi Dosaka-Akita
- Department of Medical Oncology, Faculty of Medicine & Graduate School of Medicine, Hokkaido University, Hokkaido
| | | | - Hiroshi Nishihara
- Genomics Unit, Keio Cancer Center, Keio University School of Medicine, Tokyo
| | - Naoko Sueoka-Aragane
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Faculty of Medicine, Saga University, Saga
| | - Shinichi Toyooka
- Department of General Thoracic Surgery and Breast and Endocrinological Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Science, Okayama
| | | | - Ukihide Tateishi
- Department of Diagnostic Radiology, Tokyo Medical and Dental University, Tokyo, Japan
| | | | - Sadakatsu Ikeda
- Center for Innovative Cancer Treatment
- Moores Cancer Center, University of California, San Diego, CA
| |
Collapse
|
229
|
Madison R, Schrock AB, Castellanos E, Gregg JP, Snider J, Ali SM, Miller VA, Singal G, Alexander BM, Venstrom JM, Chung JH. Retrospective analysis of real-world data to determine clinical outcomes of patients with advanced non-small cell lung cancer following cell-free circulating tumor DNA genomic profiling. Lung Cancer 2020; 148:69-78. [PMID: 32823229 DOI: 10.1016/j.lungcan.2020.07.033] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 07/12/2020] [Accepted: 07/27/2020] [Indexed: 12/15/2022]
Abstract
OBJECTIVES Liquid biopsy and comprehensive genomic profiling (CGP) of circulating tumor DNA (ctDNA) are increasingly used for detection of targetable genomic alterations (GA) in non-small cell lung cancer (NSCLC). To examine the clinical outcomes for patients following CGP using liquid biopsy versus tissue biopsy, receipt of matched targeted therapy post-CGP and associated outcomes were evaluated in the real-world setting. METHODS 6491 patients with NSCLC and liquid biopsy (N = 937 tests) and/or tissue (N = 5582 tests) CGP were included in a de-identified commercial clinico-genomic database. Targetable GAs included National Comprehensive Cancer Network NSCLC guideline biomarkers. Clinical characteristics, real-world progression, and real-world response (rwR) were obtained via technology-enabled abstraction of clinician notes and radiology/pathology reports. RESULTS At the time of liquid biopsy CGP, 53% (496/937) of patients were documented to have received ≥1 line of prior therapy (tissue CGP: 13%, 735/5582). 90% (832/928) of liquid biopsy cases had evidence of ctDNA. A targetable GA was detected in 20% (188/937) of liquid biopsy and 22% (1215/5582) of tissue CGP cases. Use of matched targeted therapy overall was similar post-liquid biopsy or post-tissue CGP but varied considerably across emerging (25%, 79/317) versus standard of care (SOC) (74%, 475/640) GA. Real-world-progression free survival for patients receiving SOC first line matched targeted therapy administered following liquid biopsy (n = 33) and tissue (n = 229) CGP were similar (13.8 vs 10.6 months; aHR = 0.68 [0.36-1.26]). Among patients evaluated for rwR, overall response rate (partial/complete response) to matched targeted therapy post-liquid biopsy CGP was 75% (39/52) versus 66% post-tissue CGP (254/385, P = 0.51). CONCLUSION Retrospective analysis of real-world clinico-genomic data demonstrated that clinical outcomes on matched targeted therapy were similar following liquid biopsy and tissue CGP in NSCLC, which suggests routine clinical use of liquid biopsy CGP can reliably guide therapy selection.
Collapse
|
230
|
Colomer R, Mondejar R, Romero-Laorden N, Alfranca A, Sanchez-Madrid F, Quintela-Fandino M. When should we order a next generation sequencing test in a patient with cancer? EClinicalMedicine 2020; 25:100487. [PMID: 32775973 PMCID: PMC7397394 DOI: 10.1016/j.eclinm.2020.100487] [Citation(s) in RCA: 96] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 07/12/2020] [Accepted: 07/16/2020] [Indexed: 12/31/2022] Open
Abstract
Technical advances in genome sequencing and the implementation of next-generation sequencing (NGS) in clinical oncology have paved the way for individualizing cancer patient therapy based on molecular profiles. When and how to use NGS testing in the clinic is at present an unsolved issue, although new research results provide evidence favoring this approach in some types of advanced cancer. Clinical research is evolving rapidly, from basket and umbrella trials to adaptative design precision oncology clinical studies, and genomic and molecular data often displace the classical clinical validation procedures of biomarkers. In this context, physicians must be aware of the clinical evidence behind these new biomarkers and NGS tests available, in order to use them in the right moment, and with a critical point of view. This review will present the status of currently available targeted drugs that can be effective based on actionable molecular alterations, and the NGS tests that are currently available, offering a practical guide for the application of Clinical Precision Oncology in the real world routine practice.
Collapse
Affiliation(s)
- Ramon Colomer
- Departament of Medicine, Universidad Autónoma de Madrid (UAM), Spain
- Medical Oncology Division, Hospital Universitario La Princesa, Madrid, Spain
- Endowed Chair of Personalised Precision Medicine, Universidad Autónoma de Madrid (UAM)-Fundación Instituto Roche, Spain
- Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid, Spain
| | - Rebeca Mondejar
- Departament of Medicine, Universidad Autónoma de Madrid (UAM), Spain
- Medical Oncology Division, Hospital Universitario La Princesa, Madrid, Spain
- Endowed Chair of Personalised Precision Medicine, Universidad Autónoma de Madrid (UAM)-Fundación Instituto Roche, Spain
| | - Nuria Romero-Laorden
- Medical Oncology Division, Hospital Universitario La Princesa, Madrid, Spain
- Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid, Spain
| | | | - Francisco Sanchez-Madrid
- Departament of Medicine, Universidad Autónoma de Madrid (UAM), Spain
- Endowed Chair of Personalised Precision Medicine, Universidad Autónoma de Madrid (UAM)-Fundación Instituto Roche, Spain
- Immunology Division, Hospital Universitario La Princesa, Madrid, Spain
| | - Miguel Quintela-Fandino
- Departament of Medicine, Universidad Autónoma de Madrid (UAM), Spain
- Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid, Spain
- Medical Oncology Division, Hospital Universitario Quirón, Pozuelo de Alarcón – Madrid, Spain
| |
Collapse
|
231
|
Yee LM, McShane LM, Freidlin B, Mooney MM, Korn EL. Biostatistical and Logistical Considerations in the Development of Basket and Umbrella Clinical Trials. Cancer J 2020; 25:254-263. [PMID: 31335389 PMCID: PMC6658126 DOI: 10.1097/ppo.0000000000000384] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Oncology clinical trials are undergoing transformation to evaluate targeted therapies addressing a wider variety of biologically defined cancer subgroups. Multiarm basket and umbrella trials conducted under master protocols have become more prominent mechanisms for the clinical evaluation of promising new biologically driven anticancer therapies that are integral to precision oncology medicine. These new trial designs permit efficient clinical evaluation of multiple therapies in a variety of histologically and biologically defined cancers. These complex trials require extensive planning and attention to many factors, including choice of biomarker assay platform, mechanism for processing clinicopathologic and biomarker data to assign patients to substudies, and statistical design, monitoring, and analysis of substudies. Trial teams have expanded to include expertise in the interface between biology, clinical oncology, bioinformatics, and statistics. Strategies for the design, conduct, and analysis of these complex trials will continue to evolve to meet new challenges and opportunities in precision oncology medicine.
Collapse
Affiliation(s)
- Laura M. Yee
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD
| | - Lisa M. McShane
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD
| | - Boris Freidlin
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD
| | - Margaret M. Mooney
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD
| | - Edward L. Korn
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD
| |
Collapse
|
232
|
Abstract
The ability of next-generation sequencing (NGS) to comprehensively assess the molecular profile of a tumor specimen has transformed the clinical testing landscape in oncology. Accordingly, recent years have seen broad uptake of clinical NGS to inform cancer patient management. However, significant challenges remain. The annotation and clinical interpretation of variants identified by NGS tests often require rigorous review and may vary between laboratories. While a clearer regulatory path has emerged, reimbursement for NGS tests remains a subject of continuing debate. Basket clinical studies such as the National Cancer Institute Molecular Analysis of Therapy Choice are evaluating the degree to which matching of a targeted therapy to tumor molecular profile by NGS can be applied independently of tissue histology. Newer applications of NGS such as for circulating tumor DNA testing and to identify novel RNA fusion driver events continue to expand its clinical utility.
Collapse
Affiliation(s)
- Chris A. Karlovich
- Leidos Biomedical Research Inc. Molecular Characterization Laboratory, Frederick National Laboratory for Cancer Research, 1050 Boyles Street, 459/108, Frederick, MD 21702
| | - P. Mickey Williams
- Leidos Biomedical Research Inc. Molecular Characterization Laboratory, Frederick National Laboratory for Cancer Research, 1050 Boyles Street, 459/108, Frederick, MD 21702
| |
Collapse
|
233
|
Gower A, Golestany B, Gong J, Singhi AD, Hendifar AE. Novel ALK Fusion, PPFIBP1-ALK, in Pancreatic Ductal Adenocarcinoma Responsive to Alectinib and Lorlatinib. JCO Precis Oncol 2020; 4:PO.19.00365. [PMID: 32923899 PMCID: PMC7446504 DOI: 10.1200/po.19.00365] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/15/2020] [Indexed: 12/14/2022] Open
Affiliation(s)
- Arjan Gower
- Department of Medicine, Cedars Sinai Medical Center, Los Angeles, CA
| | - Barry Golestany
- Department of Radiology, Cedars Sinai Medical Center, Los Angeles, CA
| | - Jun Gong
- Division of Hematology and Oncology, Cedars Sinai Medical Center, Los Angeles, CA
| | - Aatur D. Singhi
- Department of Pathology, University of Pittsburgh Medical Center, Pittsburgh, PA
| | | |
Collapse
|
234
|
Martini G, Dienstmann R, Ros J, Baraibar I, Cuadra-Urteaga JL, Salva F, Ciardiello D, Mulet N, Argiles G, Tabernero J, Elez E. Molecular subtypes and the evolution of treatment management in metastatic colorectal cancer. Ther Adv Med Oncol 2020; 12:1758835920936089. [PMID: 32782486 PMCID: PMC7383645 DOI: 10.1177/1758835920936089] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 05/22/2020] [Indexed: 12/22/2022] Open
Abstract
Colorectal cancer (CRC) is a heterogeneous disease representing a therapeutic challenge, which is further complicated by the common occurrence of several molecular alterations that confer resistance to standard chemotherapy and targeted agents. Mechanisms of resistance have been identified at multiple levels in the epidermal growth factor receptor (EGFR) pathway, including mutations in KRAS, NRAS, and BRAF V600E, and in the HER2 and MET receptors. These alterations represent oncogenic drivers that may co-exist in the same tumor with other primary and acquired alterations via a clonal selection process. Other molecular alterations include DNA damage repair mechanisms and rare kinase fusions, potentially offering a rationale for new therapeutic strategies. In recent years, genomic analysis has been expanded by a more complex study of epigenomic, transcriptomic, and microenvironment features. The Consensus Molecular Subtype (CMS) classification describes four CRC subtypes with distinct biological characteristics that show prognostic and potential predictive value in the clinical setting. Here, we review the panorama of actionable targets in CRC, and the developments in more recent molecular tests, such as liquid biopsy analysis, which are increasingly offering clinicians a means of ensuring optimal tailored treatments for patients with metastatic CRC according to their evolving molecular profile and treatment history.
Collapse
Affiliation(s)
- Giulia Martini
- Università della Campania L. Vanvitelli, Naples
- Vall d’Hebron Institute of Oncology, P/ Vall D’Hebron 119-121, Barcelona, 08035, Spain
| | | | - Javier Ros
- Vall d’Hebron Hospital, Barcelona, Catalunya, Spain
| | | | | | | | - Davide Ciardiello
- Università della Campania L. Vanvitelli, Naples
- Vall d’Hebron Hospital, Barcelona, Catalunya, Spain
| | - Nuria Mulet
- Vall d’Hebron Hospital, Barcelona, Catalunya, Spain
| | | | | | - Elena Elez
- Vall D’Hebron Institute of Oncology P/Vall D’Hebron 119-121, Barcelona, 08035 Spain
| |
Collapse
|
235
|
Deligiorgi MV, Trafalis DT. Repurposing denosumab in lung cancer beyond counteracting the skeletal related events: an intriguing perspective. Expert Opin Biol Ther 2020; 20:1331-1346. [PMID: 32658547 DOI: 10.1080/14712598.2020.1790522] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
INTRODUCTION Repurposing denosumab in lung cancer therapeutics capitalizes on its well-established role in preventing the skeletal related events (SREs) and its emerging, yet elusive, bone-independent role, assigned to inhibit the contribution of RANKL to cancer initiation and progression. AREAS COVERED The present review presents the available preclinical and clinical data indicating that denosumab may provide survival benefit to lung cancer patients beyond the counteraction of SREs. EXPERT OPINION Despite the preliminary data heralding the potential of denosumab to increase overall survival in lung cancer, the embracement of this strategy in clinical practice cannot be advocated until large randomized clinical trials consolidate its safety and efficacy. Given the improvement of lung cancer prognosis ascribed to revolutionary targeted treatment agents, the possibility of denosumab-related increased risk of second primary malignancies merits further evaluation. Many challenges in endorsing denosumab as a strategy to treat lung cancer beyond SREs prevention are pending counteraction, including: (i) patient selection guided by validated predictive and prognostic biomarkers; (ii) assessment of long-term outcomes; (iii) evaluation of benefit-risk ratio; (iv) translational research; (v) combination of denosumab with other targeted therapies; (vi) integration of genomic biomarkers, immune-related biomarkers, and biomarkers of active RANKL pathway to guide the decision-making process.
Collapse
Affiliation(s)
- Maria V Deligiorgi
- Department of Pharmacology - Clinical Pharmacology Unit, National and Kapodistrian University of Athens, Faculty of Medicine , Athens, Greece
| | - Dimitrios T Trafalis
- Department of Pharmacology - Clinical Pharmacology Unit, National and Kapodistrian University of Athens, Faculty of Medicine , Athens, Greece
| |
Collapse
|
236
|
Metro G, Baglivo S, Moretti R, Bellezza G, Sidoni A, Roila F. Is There a Role for Multiple Lines of Anti-HER2 Therapies Administered Beyond Progression in HER2-Mutated Non-Small Cell Lung Cancer? A Case Report and Literature Review. Oncol Ther 2020; 8:341-350. [PMID: 32700047 PMCID: PMC7683654 DOI: 10.1007/s40487-020-00121-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Indexed: 12/17/2022] Open
Abstract
Oncogene-addicted non-small cell lung cancer (NSCLC) comprises a number of distinct disease subtypes, each of which is characterised by druggable genetic alterations. Among them, the receptor tyrosine kinase protein human epidermal receptor 2 (HER2) is occasionally found deregulated via gene mutation and/or amplification and/or protein overexpression. HER2 mutation, in particular, is a relatively rare condition which occurs in 1-4% of NSCLC patients, especially in those with adenocarcinoma histology and a never/light smoking history. However, the clinical relevance of a HER2 mutation in NSCLC relies on the fact that this genetic alteration has been associated with sensitivity to anti-HER2 therapies such as the monoclonal antibody trastuzumab or the pan-HER-tyrosine kinase inhibitor poziotinib. Here we describe the case of a NSCLC patient with an activating exon 20 G776VinsC mutation in the HER2 gene who responded well to multiple lines of trastuzumab-based therapies administered beyond progression and poziotinib given sequentially. In this specific case, the discovery of a druggable genetic alteration such as a mutation in the HER2 gene allowed for long-term control of the disease through the use of highly effective anti-HER2 therapies.
Collapse
Affiliation(s)
- Giulio Metro
- Medical Oncology, Santa Maria Della Misericordia Hospital, Azienda Ospedaliera Di Perugia, Perugia, Italy.
| | - Sara Baglivo
- Laboratory of Oncology, Medical Oncology, Santa Maria Della Misericordia Hospital, Azienda Ospedaliera Di Perugia, Perugia, Italy
| | - Riccardo Moretti
- Department of Radiology, Santa Maria Della Misericordia Hospital, Azienda Ospedaliera Di Perugia, Perugia, Italy
| | - Guido Bellezza
- Division of Pathology and Histology, Department of Experimental Medicine, University of Perugia Medical School, Perugia, Italy
| | - Angelo Sidoni
- Division of Pathology and Histology, Department of Experimental Medicine, University of Perugia Medical School, Perugia, Italy
| | - Fausto Roila
- Medical Oncology, Santa Maria Della Misericordia Hospital, Azienda Ospedaliera Di Perugia, Perugia, Italy
| |
Collapse
|
237
|
Wakai T, Nagahashi M, Shimada Y, Prasoon P, Sakata J. Genetic analysis in the clinical management of biliary tract cancer. Ann Gastroenterol Surg 2020; 4:316-323. [PMID: 32724874 PMCID: PMC7382432 DOI: 10.1002/ags3.12334] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 02/24/2020] [Accepted: 03/12/2020] [Indexed: 12/15/2022] Open
Abstract
Biliary tract cancer (BTC) is clinically and pathologically heterogeneous and responds inadequately to treatment. A small section of patients develop resectable disease, although the relapse rates are high; the benefits of adjuvant capecitabine chemotherapy for BTC are now understood, and gemcitabine-based combination chemotherapy is the first line of therapeutic strategy for BTC; however, alternative therapy for BTC is not known. Genomic profiling can provide detailed information regarding the carcinogenesis, identification, and therapy for BTC. Currently, confirmed restorative targets for BTC are lacking. In this review, we aimed to analyze the preclinical and clinical implications of a spectrum of genomic alterations associated with new potentially remedial targets. We focused on eight draggable genes for BTC, which were described as having evidence of therapeutic impact (evidence level 2A-3B) based on the clinical practice guidance for next-generation sequencing in cancer diagnosis and treatment; these include ERBB2, NTRK1, RNF43, CDK6, CDKN2B, FGFR2, IDH1, and IDH2. Moreover, some of the BTC present microsatellite instability, hypermutation, and germline variants, which we also reviewed. Finally, we discussed the therapeutic options based on the next-generation sequencing findings in BTC. Studies have demonstrated that BTC includes subgroups with individually distinct driver mutations, most of which will be targeted with new treatment plans.
Collapse
Affiliation(s)
- Toshifumi Wakai
- Division of Digestive and General SurgeryNiigata University Graduate School of Medical and Dental SciencesNiigataJapan
| | - Masayuki Nagahashi
- Division of Digestive and General SurgeryNiigata University Graduate School of Medical and Dental SciencesNiigataJapan
| | - Yoshifumi Shimada
- Division of Digestive and General SurgeryNiigata University Graduate School of Medical and Dental SciencesNiigataJapan
| | - Pankaj Prasoon
- Division of Digestive and General SurgeryNiigata University Graduate School of Medical and Dental SciencesNiigataJapan
| | - Jun Sakata
- Division of Digestive and General SurgeryNiigata University Graduate School of Medical and Dental SciencesNiigataJapan
| |
Collapse
|
238
|
Baraibar I, Ros J, Mulet N, Salvà F, Argilés G, Martini G, Cuadra JL, Sardo E, Ciardiello D, Tabernero J, Élez E. Incorporating traditional and emerging biomarkers in the clinical management of metastatic colorectal cancer: an update. Expert Rev Mol Diagn 2020; 20:653-664. [PMID: 32552041 DOI: 10.1080/14737159.2020.1782194] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
INTRODUCTION Molecular profiling has led to significantly longer survival in metastatic colorectal cancer (mCRC) patients. Clinical guidelines recommend testing for KRAS/NRAS, BRAF and MSI status, and new biomarkers such as HER2 amplification and NTRK fusions have emerged more recently in refractory CRC, supported by overwhelming clinical relevance. These biomarkers can guide treatment management to improve clinical outcomes in these patients. AREAS COVERED Preclinical and clinical data over the last decade were reviewed for known and novel biomarkers with clinical implications in refractory CRC. Molecular alterations are described for classic and novel biomarkers, and data for completed and ongoing studies with targeted and immunotherapies are presented. EXPERT OPINION Use of targeted therapies based on biomarker testing in CRC has enabled impressive improvements in clinical outcomes in refractory patients. BRAF, MSI, NRAS and KRAS should be tested upfront in all patients given their indisputable therapeutic implications. Other molecular alterations such as HER2 and NTRK are emerging. Testing for these alterations may further improve outcomes for refractory CRC patients. Nonetheless, many key aspects remain to be defined including the optimal timing and technique for testing, the most adequate panel, and whether all patients should be tested for all alterations.
Collapse
Affiliation(s)
- Iosune Baraibar
- Department of Medical Oncology, Vall d'Hebron University Hospital , Barcelona, Spain.,Department of Medical Oncology, Vall d'Hebron Institute of Oncology (VHIO) , Barcelona, Spain
| | - Javier Ros
- Department of Medical Oncology, Vall d'Hebron University Hospital , Barcelona, Spain.,Department of Medical Oncology, Vall d'Hebron Institute of Oncology (VHIO) , Barcelona, Spain
| | - Nuria Mulet
- Department of Medical Oncology, Vall d'Hebron Institute of Oncology (VHIO) , Barcelona, Spain.,Department of Medical Oncology, Institut Català D' oncologia-IDIBELL , Barcelona, Spain
| | - Francesc Salvà
- Department of Medical Oncology, Vall d'Hebron University Hospital , Barcelona, Spain.,Department of Medical Oncology, Vall d'Hebron Institute of Oncology (VHIO) , Barcelona, Spain
| | - Guillem Argilés
- Department of Medical Oncology, Vall d'Hebron University Hospital , Barcelona, Spain.,Department of Medical Oncology, Vall d'Hebron Institute of Oncology (VHIO) , Barcelona, Spain
| | - Giulia Martini
- Department of Medical Oncology, Vall d'Hebron Institute of Oncology (VHIO) , Barcelona, Spain.,Dipartimento di Medicina di Precisione, Università Degli Studi Della Campania Luigi Vanvitelli , Naples, Italy
| | | | - Emilia Sardo
- Department of Medical Oncology, Vall d'Hebron University Hospital , Barcelona, Spain
| | - Davide Ciardiello
- Department of Medical Oncology, Vall d'Hebron Institute of Oncology (VHIO) , Barcelona, Spain.,Dipartimento di Medicina di Precisione, Università Degli Studi Della Campania Luigi Vanvitelli , Naples, Italy
| | - Josep Tabernero
- Department of Medical Oncology, Vall d'Hebron University Hospital , Barcelona, Spain.,Department of Medical Oncology, Vall d'Hebron Institute of Oncology (VHIO) , Barcelona, Spain
| | - Elena Élez
- Department of Medical Oncology, Vall d'Hebron University Hospital , Barcelona, Spain.,Department of Medical Oncology, Vall d'Hebron Institute of Oncology (VHIO) , Barcelona, Spain
| |
Collapse
|
239
|
Meyer EL, Mesenbrink P, Dunger-Baldauf C, Fülle HJ, Glimm E, Li Y, Posch M, König F. The Evolution of Master Protocol Clinical Trial Designs: A Systematic Literature Review. Clin Ther 2020; 42:1330-1360. [PMID: 32622783 DOI: 10.1016/j.clinthera.2020.05.010] [Citation(s) in RCA: 76] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 04/10/2020] [Accepted: 05/11/2020] [Indexed: 02/07/2023]
Abstract
PURPOSE Recent years have seen a change in the way that clinical trials are being conducted. There has been a rise of designs more flexible than traditional adaptive and group sequential trials which allow the investigation of multiple substudies with possibly different objectives, interventions, and subgroups conducted within an overall trial structure, summarized by the term master protocol. This review aims to identify existing master protocol studies and summarize their characteristics. The review also identifies articles relevant to the design of master protocol trials, such as proposed trial designs and related methods. METHODS We conducted a comprehensive systematic search to review current literature on master protocol trials from a design and analysis perspective, focusing on platform trials and considering basket and umbrella trials. Articles were included regardless of statistical complexity and classified as reviews related to planned or conducted trials, trial designs, or statistical methods. The results of the literature search are reported, and some features of the identified articles are summarized. FINDINGS Most of the trials using master protocols were designed as single-arm (n = 29/50), Phase II trials (n = 32/50) in oncology (n = 42/50) using a binary endpoint (n = 26/50) and frequentist decision rules (n = 37/50). We observed an exponential increase in publications in this domain during the last few years in both planned and conducted trials, as well as relevant methods, which we assume has not yet reached its peak. Although many operational and statistical challenges associated with such trials remain, the general consensus seems to be that master protocols provide potentially enormous advantages in efficiency and flexibility of clinical drug development. IMPLICATIONS Master protocol trials and especially platform trials have the potential to revolutionize clinical drug development if the methodologic and operational challenges can be overcome.
Collapse
Affiliation(s)
- Elias Laurin Meyer
- Center for Medical Statistics, Informatics, and Intelligent Systems, Medical University of Vienna, Vienna, Austria
| | | | | | | | | | - Yuhan Li
- Novartis Pharmaceuticals Corporation, East Hanover, NJ, USA
| | - Martin Posch
- Center for Medical Statistics, Informatics, and Intelligent Systems, Medical University of Vienna, Vienna, Austria
| | - Franz König
- Center for Medical Statistics, Informatics, and Intelligent Systems, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
240
|
Jeong ISD, Moyers J, Thung I, Thinn MM. Combination Chemohormonal Therapy in Metastatic Salivary Duct Carcinoma. AMERICAN JOURNAL OF CASE REPORTS 2020; 21:e925181. [PMID: 32601266 PMCID: PMC7347037 DOI: 10.12659/ajcr.925181] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Patient: Male, 68-year-old Final Diagnosis: Salivary duct carcinoma Symptoms: Bone pain • dyspnea Medication:— Clinical Procedure: Chemotherapy Specialty: Oncology
Collapse
Affiliation(s)
| | - Justin Moyers
- Division of Hematology and Oncology, Department of Internal Medicine, Loma Linda University Medical Center, Loma Linda, CA, USA
| | - Irene Thung
- Department of Pathology, Veterans Affairs Loma Linda Healthcare System, Loma Linda, CA, USA
| | - Mie Mie Thinn
- Department of Hematology and Oncology, Veterans Affairs Loma Linda Healthcare System, Loma Linda, CA, USA
| |
Collapse
|
241
|
Wang X, Hu W, Xie L. Response to Anti-HER2-Based Treatment in a Patient with Bladder Adenocarcinoma Harboring HER2 Amplification and S310F Mutation Discovered by Next-Generation Sequencing: A Case Report. Onco Targets Ther 2020; 13:4249-4255. [PMID: 32547059 PMCID: PMC7244354 DOI: 10.2147/ott.s247515] [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: 01/28/2020] [Accepted: 04/19/2020] [Indexed: 11/23/2022] Open
Abstract
Purpose HER2 overexpression has been identified in approximately 14% of bladder adenocarcinomas. However, until now, there has been no approved standard targeted therapy for bladder adenocarcinoma patients harboring HER2 genetic alteration. Case Presentation We presented a case of a 64-year-old man who was diagnosed with bladder adenocarcinoma, and lung metastasis was confirmed less than one year after initial bladder surgery. The patient received systemic chemotherapy and antiangiogenetic treatment, but the tumor continued to progress. The patient underwent next-generation sequencing (NGS) to seek potential treatment opportunities. HER2 amplification, approximately 7 times, was discovered together with the S310F mutation (mutant abundance 90%). The patient then received late-line treatment with trastuzumab and albumin-bound paclitaxel. A partial response was confirmed two months later. Trastuzumab-based therapy was continued for 8 cycles, and the progression-free survival period was 6 months. NGS was performed on a rebiopsy, and the result showed no amplification of HER2, and the S310F mutant abundance was reduced to 27.9%. Conclusion This is the first case report describing a bladder adenocarcinoma patient harboring HER2 amplification who responded to trastuzumab. NGS is of great potential in the selection of bladder adenocarcinoma patients suitable for anti-HER2 therapy. The genetic change after treatment also implied possible mechanisms of resistance to trastuzumab-based therapy, which requires more investigation.
Collapse
Affiliation(s)
- Xiaolu Wang
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University, Clinical Cancer Institute of Nanjing University, Nanjing, People's Republic of China
| | - Wenjing Hu
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University, Clinical Cancer Institute of Nanjing University, Nanjing, People's Republic of China
| | - Li Xie
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University, Clinical Cancer Institute of Nanjing University, Nanjing, People's Republic of China
| |
Collapse
|
242
|
Rational Cancer Treatment Combinations: An Urgent Clinical Need. Mol Cell 2020; 78:1002-1018. [DOI: 10.1016/j.molcel.2020.05.031] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 05/20/2020] [Accepted: 05/21/2020] [Indexed: 02/07/2023]
|
243
|
Tsimberidou AM, Fountzilas E, Nikanjam M, Kurzrock R. Review of precision cancer medicine: Evolution of the treatment paradigm. Cancer Treat Rev 2020; 86:102019. [PMID: 32251926 PMCID: PMC7272286 DOI: 10.1016/j.ctrv.2020.102019] [Citation(s) in RCA: 392] [Impact Index Per Article: 78.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 03/22/2020] [Accepted: 03/24/2020] [Indexed: 12/15/2022]
Abstract
In recent years, biotechnological breakthroughs have led to identification of complex and unique biologic features associated with carcinogenesis. Tumor and cell-free DNA profiling, immune markers, and proteomic and RNA analyses are used to identify these characteristics for optimization of anticancer therapy in individual patients. Consequently, clinical trials have evolved, shifting from tumor type-centered to gene-directed, histology-agnostic, with innovative adaptive design tailored to biomarker profiling with the goal to improve treatment outcomes. A plethora of precision medicine trials have been conducted. The majority of these trials demonstrated that matched therapy is associated with superior outcomes compared to non-matched therapy across tumor types and in specific cancers. To improve the implementation of precision medicine, this approach should be used early in the course of the disease, and patients should have complete tumor profiling and access to effective matched therapy. To overcome the complexity of tumor biology, clinical trials with combinations of gene-targeted therapy with immune-targeted approaches (e.g., checkpoint blockade, personalized vaccines and/or chimeric antigen receptor T-cells), hormonal therapy, chemotherapy and/or novel agents should be considered. These studies should target dynamic changes in tumor biologic abnormalities, eliminating minimal residual disease, and eradicating significant subclones that confer resistance to treatment. Mining and expansion of real-world data, facilitated by the use of advanced computer data processing capabilities, may contribute to validation of information to predict new applications for medicines. In this review, we summarize the clinical trials and discuss challenges and opportunities to accelerate the implementation of precision oncology.
Collapse
Affiliation(s)
- Apostolia M Tsimberidou
- The University of Texas MD Anderson Cancer Center, Department of Investigational Cancer Therapeutics, Houston, TX.
| | - Elena Fountzilas
- Department of Medical Oncology, Euromedica General Clinic, Thessaloniki, Greece
| | - Mina Nikanjam
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, UC San Diego Moores Cancer Center, San Diego, CA, USA
| | - Razelle Kurzrock
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, UC San Diego Moores Cancer Center, San Diego, CA, USA
| |
Collapse
|
244
|
|
245
|
C. Huynh J, Schwab E, Ji J, Kim E, Joseph A, Hendifar A, Cho M, Gong J. Recent Advances in Targeted Therapies for Advanced Gastrointestinal Malignancies. Cancers (Basel) 2020; 12:E1168. [PMID: 32384640 PMCID: PMC7281439 DOI: 10.3390/cancers12051168] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Revised: 04/20/2020] [Accepted: 04/30/2020] [Indexed: 12/19/2022] Open
Abstract
The treatment of advanced gastrointestinal (GI) cancers has become increasingly molecularly driven. Molecular profiling for HER2 and PD-L1 status is standard for metastatic gastroesophageal (GEJ) cancers to predict benefits from trastuzumab (HER2-targeted therapy) and pembrolizumab (anti-PD-1 therapy), while extended RAS and BRAF testing is standard in metastatic colorectal cancer to predict benefits from epidermal growth factor receptor (EGFR)-targeted therapies. Mismatch repair (MMR) or microsatellite instability (MSI) testing is standard for all advanced GI cancers to predict benefits from pembrolizumab and in metastatic colorectal cancer, nivolumab with or without ipilimumab. Here we review recent seminal trials that have further advanced targeted therapies in these cancers including Poly (adenosine diphosphate-ribose) polymerases (PARP) inhibition in pancreas cancer, BRAF inhibition in colon cancer, and isocitrate dehydrogenase (IDH) and fibroblast growth factor receptor (FGFR) inhibition in biliary tract cancer. Targeted therapies in GI malignancies constitute an integral component of the treatment paradigm in these advanced cancers and have widely established the need for standard molecular profiling to identify candidates.
Collapse
Affiliation(s)
- Jasmine C. Huynh
- Hematology Oncology, University of California, Davis, Sacramento, CA 95817, USA; (E.K.); (A.J.); (M.C.)
| | - Erin Schwab
- Hematology Oncology, University of California, Davis, Sacramento, CA 95817, USA; (E.K.); (A.J.); (M.C.)
| | - Jingran Ji
- Internal Medicine, University of California, Davis, Sacramento, CA 95817, USA;
| | - Edward Kim
- Hematology Oncology, University of California, Davis, Sacramento, CA 95817, USA; (E.K.); (A.J.); (M.C.)
| | - Anjali Joseph
- Hematology Oncology, University of California, Davis, Sacramento, CA 95817, USA; (E.K.); (A.J.); (M.C.)
| | - Andrew Hendifar
- Hematology Oncology, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (A.H.); (J.G.)
| | - May Cho
- Hematology Oncology, University of California, Davis, Sacramento, CA 95817, USA; (E.K.); (A.J.); (M.C.)
| | - Jun Gong
- Hematology Oncology, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (A.H.); (J.G.)
| |
Collapse
|
246
|
Zhang H, Moisini I, Ajabnoor RM, Turner BM, Hicks DG. Applying the New Guidelines of HER2 Testing in Breast Cancer. Curr Oncol Rep 2020; 22:51. [PMID: 32346807 DOI: 10.1007/s11912-020-0901-4] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
PURPOSE OF REVIEW The human epidermal growth factor receptor 2 (HER2) is an important prognostic and predictive biomarker in the breast cancer. The American Society of Clinical Oncology/College of American Pathology (ASCO/CAP) has published HER2 testing guidelines in breast cancer. We herein reviewed the HER2 testing guidelines in breast cancer with a focus on the application of the current guidelines. RECENT FINDINGS The continual investigation of HER2 testing in breast cancer has resulted in updates in the HER2 testing guidelines. The current guidelines focus on the uncommon clinical scenarios and emphasize the coordination between immunohistochemistry and in situ hybridization results, in an effort to improve clarity and accuracy. The ASCO/CAP guidelines provide valuable recommendations to ensure the accurate evaluation of HER2 status in breast cancer patients through standardization. Additional studies, particularly those with long-term outcome data are still needed to validate the guideline recommendations, especially the uncommon cases.
Collapse
Affiliation(s)
- Huina Zhang
- Department of Pathology, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Ioana Moisini
- Department of Pathology, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Rana M Ajabnoor
- Department of Pathology, Faculty of medicine, King Abdulaziz University, Jeddah, 21589, Kingdom of Saudi Arabia
| | - Bradley M Turner
- Department of Pathology, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - David G Hicks
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, 601 Elmwood Avenue, Box 626, Rochester, NY, 14642, USA.
| |
Collapse
|
247
|
Chung YC, Chiu HH, Wei WC, Chang KJ, Chao WT. Application of trastuzumab emtansine in HER-2-positive and KRAS/BRAF-mutated colon cancer cells. Eur J Clin Invest 2020; 50:e13255. [PMID: 32350854 DOI: 10.1111/eci.13255] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Revised: 04/23/2020] [Accepted: 04/26/2020] [Indexed: 12/13/2022]
Abstract
BACKGROUND Trastuzumab emtansine (T-DM1) is an antibody-drug conjugate (ADC) for the treatment of human epidermal growth factor receptor 2 (HER-2)-positive breast cancer. T-DM1 is based on the trastuzumab antibody and delivers a toxic agent into breast cancer cells through endocytic mechanism. This study evaluated whether T-DM1 can be used in HER-2-positive colon cancer cells which harbour KRAS/ BRAF mutation with limited treatment. MATERIALS AND METHODS LS174T and HT-29 which are KRAS and BRAF mutant HER-2-positive colon cancer cells were used in this study. Cells were first treated with T-DM1; cetuximab and trastuzumab were applied for comparison, the effect of drug sensitivity was determined. Cells were then transfected with plasmid to overexpress HER-2 or the endocytic protein, caveolin-1 or furthermore pretreated with metformin to examine the effect of T-DM1 efficacy. Finally, a xenograft mouse model was used to evaluate the drug efficacy in vivo. RESULTS The results showed that T-DM1 had better inhibitory effect than cetuximab and trastuzumab on LS174T and HT-29 cells. HER-2 or caveolin-1 overexpression with plasmid in the cells to increase T-DM1 recognition or internalization can increase the sensitivity to T-DM1. When cells were pretreated with metformin, caveolin-1 expression was induced and promoted T-DM1 uptake and enhanced cell toxicity. In xenograft mouse model, combined treatment of T-DM1 and metformin had apparent inhibitory effect on subcutaneous tumour growth. CONCLUSION The results of this study suggested that T-DM1 has potential in the treatment of HER-2-positive colon cancer cells, and application of metformin has therapeutic benefits during T-DM1 treatment.
Collapse
Affiliation(s)
- Yuan-Chiang Chung
- Department of Surgery, Cheng-Ching General Hospital, Taichung, Taiwan
| | - Hsi-Hsiung Chiu
- Department of Proctology, Cheng-Ching General Hospital, Taichung, Taiwan
| | - Wan-Chen Wei
- Department of Surgery, Cheng-Ching General Hospital, Taichung, Taiwan
- Department of Life Science, Tunghai University, Taichung, Taiwan
| | - King-Jen Chang
- Department of Breast Surgery, Cardinal Tien Hospital, Taipei, Taiwan
| | - Wei-Ting Chao
- Department of Life Science, Tunghai University, Taichung, Taiwan
| |
Collapse
|
248
|
Mu Y, Yang K, Hao X, Wang Y, Wang L, Liu Y, Lin L, Li J, Xing P. Clinical Characteristics and Treatment Outcomes of 65 Patients With BRAF-Mutated Non-small Cell Lung Cancer. Front Oncol 2020; 10:603. [PMID: 32411601 PMCID: PMC7198730 DOI: 10.3389/fonc.2020.00603] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2019] [Accepted: 04/02/2020] [Indexed: 12/26/2022] Open
Abstract
BRAF mutation is an oncogenic driver gene in non-small cell lung cancer (NSCLC) with low frequency. The data of patients with NSCLC harboring BRAF mutations is rare. We conducted a retrospective multicenter study in Chinese patients with NSCLC harboring BRAF mutations between Jan 2017 and Jul 2019. A total of 65 patients treated in 22 centers were included, 54 harbored BRAF-V600E mutation and 11 had non-V600E mutations, including K601E, G469S, G469V, G469A, G596R, G466R, and T599dup. Of 18 patients with early-stage disease at diagnosis and underwent a resection, the median disease-free survival (DFS) was 43.2, 18.7, and 10.1 months of stage I, II, and IIIA patients, respectively. In 46 patients with advanced-stage disease at data cutoff, disease control rate (DCR), and progression-free survival (PFS) of first-line anti-BRAF targeted therapy was superior than chemotherapy in patients harboring BRAF-V600E mutation (DCR, 100.0 vs. 70.0%, P = 0.027; median PFS, 9.8 vs. 5.4 months, P = 0.149). Of 30 V600E-mutated patients who received anti-BRAF therapy during the course of disease, median PFS of vemurafenib, dabrafenib, and dabrafenib plus trametinib was 7.8, 5.8, and 6.0 months, respectively (P = 0.970). Median PFS were similar between V600E and non-V600E patients (5.4 vs. 5.4 months, P = 0.825) to first-line chemotherapy. Nine patients were treated with checkpoint inhibitors, with median PFS of 3.0 months. Our data demonstrated the clinical benefit of anti-BRAF targeted therapy in Chinese NSCLC patients harboring BRAF-V600E mutation. The value of immunotherapy and treatment selection among non-V600E population needs further study.
Collapse
Affiliation(s)
- Yuxin Mu
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ke Yang
- Department of Medical Oncology, Cancer Hospital of HuanXing, Beijing, China
| | - Xuezhi Hao
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yan Wang
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lin Wang
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yutao Liu
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lin Lin
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Junling Li
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Puyuan Xing
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
249
|
Gambardella V, Tarazona N, Cejalvo JM, Lombardi P, Huerta M, Roselló S, Fleitas T, Roda D, Cervantes A. Personalized Medicine: Recent Progress in Cancer Therapy. Cancers (Basel) 2020; 12:E1009. [PMID: 32325878 PMCID: PMC7226371 DOI: 10.3390/cancers12041009] [Citation(s) in RCA: 147] [Impact Index Per Article: 29.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 04/05/2020] [Accepted: 04/15/2020] [Indexed: 01/01/2023] Open
Abstract
Translational research has revolutionized how we develop new treatments for cancer patients. The change from an organ-centric concept guiding treatment choice towards deep molecular analysis, driving a personalized approach, is one of the most important advances of modern oncology. Several tools such as next generation sequencing and RNA sequencing have greatly improved the capacity to detect predictive and prognostic molecular alterations. Detection of gene mutations, amplifications, and fusions has therefore altered the history of several diseases in both a localized and metastatic setting. This shift in perspective, in which attention is focused on the specific molecular alterations of the tumor, has opened the door to personalized treatment. This situation is reflected in the increasing number of basket trials selecting specific molecular targets. Nonetheless, some weaknesses need to be addressed. The complexity of cancer cells enriched with concomitant molecular alterations complicates identification of the driver. Moreover, tumor heterogeneity could be responsible for the lack of benefit when targeted agents are used. In light of this, there is growing interest in the role of multidisciplinary committees or molecular tumor boards to try to enhance selection. The aim of this review is to critically analyze the evolution of cancer treatment towards a precision approach, underlining some recent successes and unexpected failures.
Collapse
Affiliation(s)
- Valentina Gambardella
- Department of Medical Oncology, INCLIVA Biomedical Research Institute, University of Valencia, 46010 Valencia, Spain; (V.G.); (N.T.); (J.M.C.); (M.H.); (S.R.); (T.F.)
- Instituto de Salud Carlos III, CIBERONC, 28220 Madrid, Spain
| | - Noelia Tarazona
- Department of Medical Oncology, INCLIVA Biomedical Research Institute, University of Valencia, 46010 Valencia, Spain; (V.G.); (N.T.); (J.M.C.); (M.H.); (S.R.); (T.F.)
- Instituto de Salud Carlos III, CIBERONC, 28220 Madrid, Spain
| | - Juan Miguel Cejalvo
- Department of Medical Oncology, INCLIVA Biomedical Research Institute, University of Valencia, 46010 Valencia, Spain; (V.G.); (N.T.); (J.M.C.); (M.H.); (S.R.); (T.F.)
| | - Pasquale Lombardi
- Department of Oncology, University of Turin; Candiolo Cancer Institute - FPO- IRCCS, 10060 Candiolo (TO), Italy;
| | - Marisol Huerta
- Department of Medical Oncology, INCLIVA Biomedical Research Institute, University of Valencia, 46010 Valencia, Spain; (V.G.); (N.T.); (J.M.C.); (M.H.); (S.R.); (T.F.)
| | - Susana Roselló
- Department of Medical Oncology, INCLIVA Biomedical Research Institute, University of Valencia, 46010 Valencia, Spain; (V.G.); (N.T.); (J.M.C.); (M.H.); (S.R.); (T.F.)
- Instituto de Salud Carlos III, CIBERONC, 28220 Madrid, Spain
| | - Tania Fleitas
- Department of Medical Oncology, INCLIVA Biomedical Research Institute, University of Valencia, 46010 Valencia, Spain; (V.G.); (N.T.); (J.M.C.); (M.H.); (S.R.); (T.F.)
- Instituto de Salud Carlos III, CIBERONC, 28220 Madrid, Spain
| | - Desamparados Roda
- Department of Medical Oncology, INCLIVA Biomedical Research Institute, University of Valencia, 46010 Valencia, Spain; (V.G.); (N.T.); (J.M.C.); (M.H.); (S.R.); (T.F.)
- Instituto de Salud Carlos III, CIBERONC, 28220 Madrid, Spain
| | - Andres Cervantes
- Department of Medical Oncology, INCLIVA Biomedical Research Institute, University of Valencia, 46010 Valencia, Spain; (V.G.); (N.T.); (J.M.C.); (M.H.); (S.R.); (T.F.)
- Instituto de Salud Carlos III, CIBERONC, 28220 Madrid, Spain
| |
Collapse
|
250
|
Russo A, Lopes AR, McCusker MG, Garrigues SG, Ricciardi GR, Arensmeyer KE, Scilla KA, Mehra R, Rolfo C. New Targets in Lung Cancer (Excluding EGFR, ALK, ROS1). Curr Oncol Rep 2020; 22:48. [PMID: 32296961 DOI: 10.1007/s11912-020-00909-8] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
PURPOSE OF REVIEW Over the last two decades, the identification of targetable oncogene drivers has revolutionized the therapeutic landscape of non-small cell lung cancer (NSCLC). The extraordinary progresses made in molecular biology prompted the identification of several rare molecularly defined subgroups. In this review, we will focus on the novel and emerging actionable oncogenic drivers in NSCLC. RECENT FINDINGS Recently, novel oncogene drivers emerged as promising therapeutic targets besides the well-established EGFR mutations, and ALK/ROS1 rearrangements, considerably expanding the list of potential exploitable genetic aberrations. However, the therapeutic algorithm in these patients is far less defined. The identification of uncommon oncogene drivers is reshaping the diagnostic and therapeutic approach to NSCLC. The introduction of novel highly selective inhibitors is expanding the use of targeted therapies to rare and ultra-rare subsets of patients, further increasing the therapeutic armamentarium of advanced NSCLC.
Collapse
Affiliation(s)
- Alessandro Russo
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, 22 S Greene Street Rm. N9E08, Baltimore, MD, 21201, USA.,Medical Oncology Unit, A.O. Papardo & Department of Human Pathology, University of Messina, Contrada Papardo, 98158, Messina, Italy
| | - Ana Rita Lopes
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, 22 S Greene Street Rm. N9E08, Baltimore, MD, 21201, USA.,Portuguese Institute of Oncology (IPO), Porto, Portugal
| | - Michael G McCusker
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, 22 S Greene Street Rm. N9E08, Baltimore, MD, 21201, USA
| | - Sandra Gimenez Garrigues
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, 22 S Greene Street Rm. N9E08, Baltimore, MD, 21201, USA
| | - Giuseppina R Ricciardi
- Medical Oncology Unit, A.O. Papardo & Department of Human Pathology, University of Messina, Contrada Papardo, 98158, Messina, Italy
| | - Katherine E Arensmeyer
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, 22 S Greene Street Rm. N9E08, Baltimore, MD, 21201, USA
| | - Katherine A Scilla
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, 22 S Greene Street Rm. N9E08, Baltimore, MD, 21201, USA
| | - Ranee Mehra
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, 22 S Greene Street Rm. N9E08, Baltimore, MD, 21201, USA
| | - Christian Rolfo
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, 22 S Greene Street Rm. N9E08, Baltimore, MD, 21201, USA.
| |
Collapse
|