217 results on '"Richard D. Kim"'
Search Results
2. 187 Immune checkpoint inhibitor (CPI) efficacy in gastrointestinal tumors with microsatellite-stable with high tumor mutational burden (MSS-TMB-H) compared to microsatellite instability-high (MSI-H)
- Author
-
James Yu, Robin Park, Ruoyu Miao, Iman Imanirad, Jose Laborde, Todd Knepper, Christine Walko, and Richard D Kim
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Published
- 2023
- Full Text
- View/download PDF
3. Tumor Response-speed Heterogeneity as a Novel Prognostic Factor in Patients With Metastatic Colorectal Cancer
- Author
-
Junjia Liu, Xuefeng Wang, Ibrahim H. Sahin, Iman Imanirad, Seth I. Felder, Richard D. Kim, and Hao Xie
- Subjects
Cancer Research ,Oncology - Published
- 2022
- Full Text
- View/download PDF
4. Biomarker analysis from a phase II multi-institutional study of nivolumab in patients with advanced refractory biliary tract cancer
- Author
-
Dae Won Kim, Young-chul Kim, Bence P. Kovari, Vincent Chung, Olatunji B. Alese, Bassel F. El-Rayes, Daneng Li, Wungki Park, and Richard D. Kim
- Subjects
Cancer Research ,Nivolumab ,Antineoplastic Agents, Immunological ,Biliary Tract Neoplasms ,Oncology ,Biomarkers, Tumor ,Tumor Microenvironment ,Humans ,B7-H1 Antigen - Abstract
Our previous phase II study demonstrated that nivolumab provides modest but durable clinical efficacy in patients with refractory biliary tract cancer, suggesting the significant clinical benefit of nivolumab in selected patients and the necessity of predictive biomarkers. We evaluated clinicopathological characteristics and tumour microenvironment of the patients who were enrolled the trial to identify potential biomarkers.Baseline clinicopathological characteristics and pretreatment tumour samples were collected. The obtained tumour samples were assessed for whole exome sequencing, RNA sequencing and immunohistochemistry. Their correlations with clinical outcome were analysed.Pretreatment tumour evaluation revealed PD-L1 expression on tumour, CD8 T cell infiltration and high ratio of CD8 T cell/regulatory T cell in tumour microenvironment were significantly associated with prolonged progression-free survival (PFS), while PD-1 expression on lymphocytes and CD68 macrophages infiltration in tumour microenvironment had no predictive role. Asian patients (N = 3) had improved PFS and disease control rate compared with non-Asian (N = 54). A six-gene predictive model was constructed by evaluation of total 23,550 candidate genes from RNA sequencing of baseline tumour samples using LASSO-Cox regression analysis, and high score of the six-gene prediction model was associated with prolonged PFS.This study suggests that PD-L1 expression on tumour, CD8 T cell infiltration and high ratio of CD8/regulatory T cells and six-gene expression profile in tumour microenvironment may be potential predictive biomarkers of nivolumab in biliary tract cancers. Further studies are needed to confirm these findings.
- Published
- 2022
- Full Text
- View/download PDF
5. Radioembolization with Yttrium-90 Glass Microspheres as a First-Line Treatment for Unresectable Intrahepatic Cholangiocarcinoma – A Prospective Feasibility Study
- Author
-
Bela Kis, Ravi Shridhar, Rahul Mhaskar, Marcell Gyano, Jessica M. Frakes, Ghassan El-Haddad, Junsung Choi, Richard D. Kim, and Sarah E. Hoffe
- Subjects
Radiology, Nuclear Medicine and imaging ,Cardiology and Cardiovascular Medicine - Published
- 2023
- Full Text
- View/download PDF
6. Supplementary Data from First-in-Human Phase I Study of Fisogatinib (BLU-554) Validates Aberrant FGF19 Signaling as a Driver Event in Hepatocellular Carcinoma
- Author
-
Yoon-Koo Kang, Beni B. Wolf, Hongliang Shi, Klaus P. Hoeflich, Oleg Schmidt-Kittler, Cori Ann Sherwin, Christoph Lengauer, Nancy E. Kohl, Margit Hagel, Nicolas Stransky, Meera Tugnait, Melissa Manoogian, Josep M. Llovet, Daniel H. Palmer, Jean-Francois Dufour, Chia-Jui Yen, Lynn G. Feun, Sandrine Faivre, Stephen L. Chan, Zhong-Zhe Lin, Sunil Sharma, Jung-Hwan Yoon, Andrew X. Zhu, Joerg Trojan, Vincenzo Mazzaferro, Ho-Yeong Lim, Max W. Sung, Antoine Hollebecque, Su Pin Choo, Joong-Won Park, Teresa Macarulla, Thomas Yau, Tim Meyer, Debashis Sarker, and Richard D. Kim
- Abstract
Supplement
- Published
- 2023
- Full Text
- View/download PDF
7. Data from First-in-Human Phase I Study of Fisogatinib (BLU-554) Validates Aberrant FGF19 Signaling as a Driver Event in Hepatocellular Carcinoma
- Author
-
Yoon-Koo Kang, Beni B. Wolf, Hongliang Shi, Klaus P. Hoeflich, Oleg Schmidt-Kittler, Cori Ann Sherwin, Christoph Lengauer, Nancy E. Kohl, Margit Hagel, Nicolas Stransky, Meera Tugnait, Melissa Manoogian, Josep M. Llovet, Daniel H. Palmer, Jean-Francois Dufour, Chia-Jui Yen, Lynn G. Feun, Sandrine Faivre, Stephen L. Chan, Zhong-Zhe Lin, Sunil Sharma, Jung-Hwan Yoon, Andrew X. Zhu, Joerg Trojan, Vincenzo Mazzaferro, Ho-Yeong Lim, Max W. Sung, Antoine Hollebecque, Su Pin Choo, Joong-Won Park, Teresa Macarulla, Thomas Yau, Tim Meyer, Debashis Sarker, and Richard D. Kim
- Abstract
Outcomes for patients with advanced hepatocellular carcinoma (HCC) remain poor despite recent progress in drug development. Emerging data implicate FGF19 as a potential HCC driver, suggesting its receptor, FGFR4, as a novel therapeutic target. We evaluated fisogatinib (BLU-554), a highly potent and selective oral FGFR4 inhibitor, in a phase I dose-escalation/dose-expansion study in advanced HCC using FGF19 expression measured by IHC as a biomarker for pathway activation. For dose escalation, 25 patients received 140 to 900 mg fisogatinib once daily; the maximum tolerated dose (600 mg once daily) was expanded in 81 patients. Fisogatinib was well tolerated; most adverse events were manageable, grade 1/2 gastrointestinal events, primarily diarrhea, nausea, and vomiting. Across doses, the overall response rate was 17% in FGF19-positive patients [median duration of response: 5.3 months (95% CI, 3.7–not reached)] and 0% in FGF19-negative patients. These results validate FGFR4 as a targetable driver in FGF19-positive advanced HCC.Significance:Fisogatinib elicited clinical responses in patients with tumor FGF19 overexpression in advanced HCC. These results validate the oncogenic driver role of the FGFR4 pathway in HCC and the use of FGF19 as a biomarker for patient selection.See related commentary by Subbiah and Pal, p. 1646.This article is highlighted in the In This Issue feature, p. 1631
- Published
- 2023
- Full Text
- View/download PDF
8. Tumor Response-speed Heterogeneity as a Novel Prognostic Factor in Patients With Metastatic Colorectal Cancer
- Author
-
Junjia, Liu, Xuefeng, Wang, Ibrahim H, Sahin, Iman, Imanirad, Seth I, Felder, Richard D, Kim, and Hao, Xie
- Abstract
Differential tumor response to therapy is partially attributed to tumor heterogeneity. Additional efforts are needed to identify tumor heterogeneity parameters in response to therapy that is easily applicable in clinical practice. We aimed to describe tumor response-speed heterogeneity and evaluate its prognostic value in patients with metastatic colorectal cancer.Individual patient data from Amgen (NCT00364013) and Sanofi (NCT00305188; NCT00272051) trials were retrieved from Project Data Sphere. Patients in the Amgen 5-fluorouracil, leucovorin, oxaliplatin (FOLFOX) arm were used to establish response-speed heterogeneity. Its prognostic value was subsequently validated in the Sanofi FOLFOX arms and the Amgen panitumumab+FOLFOX arm. Kaplan-Meier method and Cox proportional hazards models were used for survival analyses.Patients with high response-speed heterogeneity in the Amgen FOLFOX cohort had significantly shorter (P0.001) median progression-free survival (PFS) of 7.27 months (95% CI, 6.12-7.96 mo) and overall survival (OS) of 16.0 months (95% CI, 13.8-18.2 mo) than patients with low response-speed heterogeneity with median PFS of 9.41 months (95% CI, 8.75-10.89 mo) and OS of 22.4 months (95% CI, 20.1-26.7 mo), respectively. Tumor response-speed heterogeneity was a poor prognostic factor of shorter PFS (hazard ratio, 4.17; 95% CI, 2.49-6.99; P0.001) and shorter OS (hazard ratio, 2.57; 95% CI, 1.64-4.01; P0.001), after adjustment for other common prognostic factors. Comparable findings were found in the external validation cohorts.Tumor response-speed heterogeneity to first-line chemotherapy was a novel prognostic factor associated with early disease progression and shorter survival in patients with metastatic colorectal cancer.
- Published
- 2023
9. Metagenomics and chemotherapy‐induced nausea: A roadmap for future research
- Author
-
Christine M. Pierce, Aasha I. Hoogland, Daneng Li, Jane C. Figueiredo, Kea Turner, Taylor L. Welniak, Sylvia L. Crowder, Stacy Fischer, Elizabeth A. Lafranchise, Danielle Tometich, George M. Weinstock, Arshiya Mariam, Thi Dong Binh Tran, Jameel Muzaffar, Daniel M. Rotroff, Heather S.L. Jim, Kristen M. Carpenter, Anita Y. Kinney, Shahla Bari, Farzaneh Rastegari, Richard D. Kim, Martine Extermann, and Donna L. Berry
- Subjects
Cancer Research ,medicine.medical_specialty ,Vomiting ,business.industry ,Nausea ,Antineoplastic Agents ,Context (language use) ,Article ,Discontinuation ,Oncology ,Quality of life ,Metagenomics ,Neoplasms ,Quality of Life ,medicine ,Etiology ,Antiemetics ,Humans ,Microbiome ,medicine.symptom ,Intensive care medicine ,business - Abstract
Uncontrolled chemotherapy-induced nausea and vomiting (CINV) can reduce patients’ quality of life and may result in premature discontinuation of chemotherapy. Although nausea and vomiting are commonly grouped together, research has shown that antiemetics are clinically effective against chemotherapy-induced vomiting (CIV) but less so against chemotherapy-induced nausea (CIN). Nausea remains a problem for up to 68% of patients who are prescribed guideline-consistent antiemetics. Despite the high prevalence of CIN, relatively little is known regarding its etiology independent of CIV. In this review paper, we summarize a metagenomics approach to the study and treatment of CIN with the goal of encouraging future research. Metagenomics focuses on genetic risk factors, encompassing both human (i.e., host) and gut microbial genetic variation. Little work to date has focused on metagenomics as a putative biological mechanism of CIN. Metagenomics has the potential to be a powerful tool in advancing scientific understanding of CIN by identifying new biological pathways and intervention targets. Investigation of metagenomics in the context of well-established demographic, clinical, and patient-reported risk factors may help to identify patients at risk and facilitate prevention and management of CIN.
- Published
- 2021
- Full Text
- View/download PDF
10. Age-related disparity of survival outcomes and treatment-related adverse events in patients with metastatic colorectal cancer
- Author
-
Lingbin Meng, Ram Thapa, Maria G. Delgado, Maria F. Gomez, Rui Ji, Todd C. Knepper, Joleen M. Hubbard, Xuefeng Wang, Jennifer B. Permuth, Richard D. Kim, Damian A. Laber, and Hao Xie
- Abstract
BackgroundWhile the incidence of newly diagnosed early-onset colorectal cancer has been increasing, age-related disparity of survival outcome and treatment-related adverse events in patients with metastatic CRC (mCRC) has been inadequately studied with inconclusive findings. In this study, we aimed to evaluate such age-related disparity in this patient population.MethodsWe used individual patient data from three clinical trials (Study 1:NCT00272051, NCT 00305188 and Study 2:NCT00364013) in Project Data Sphere. All patients were diagnosed with mCRC and received first-line 5-fluorouracil and oxaliplatin. Clinical and genomic data of 763 patients with mCRC from Moffitt Cancer Center were used to assess genomic alterations and serve as an external and real-world validation cohort to evaluate overall survival (OS) disparity. Survival outcomes and treatment-related adverse events were estimated and compared in patients among three age groups: 65 years.ResultsAmong 1223 patients from previous clinical trials, 179 (14.6%) were younger than 50 years. These patients had significantly shorter progression-free survival (PFS) (HR=1.46; 95%CI=1.22–1.76;ppp=0.019), severe abdominal pain (8.4% vs 3.4% vs 3.5%,p=0.018), severe anemia (6.1% vs 1.0% vs 1.5%,pp=0.047), but significantly lower incidence of fatigue, severe diarrhea, severe fatigue, and severe neutropenia. The p=0.012), mucositis (3.6 vs 5.1 vs 5.7 weeks,p=0.051), and neutropenia (8.0 vs 9.4 vs 8.4 weeks,p=0.043), and shorter duration of mucositis (0.6 vs 0.9 vs 1.0 weeks,p=0.006). In the CTNNB1mutation (6.6% vs 3.1% vs 2.3%,p=0.047),ERBB2amplification (5.1% vs 0.6% vs 2.3%,p=0.005), andCREBBPmutation (3.1% vs 0.9% vs 0.5%,p=0.050), but lower prevalence ofBRAFmutation (7.7% vs 8.5% vs 16.7%,p=0.002).ConclusionsPatients with early-onset mCRC had worse survival outcome and unique adverse-event patterns, which could be partially attributed to distinct genomic profiles. Our findings might improve an individualized approach to chemotherapy, counseling, and management of treatment-related adverse events in this patient population.
- Published
- 2022
- Full Text
- View/download PDF
11. Immunotherapy in Biliary Tract Cancers: Where Are We?
- Author
-
Aparna Kalyan, Harshit Khosla, and Richard D. Kim
- Subjects
Cholangiocarcinoma ,Biliary Tract Neoplasms ,Bile Ducts, Intrahepatic ,Oncology ,Bile Duct Neoplasms ,Tumor Microenvironment ,Humans ,Immunologic Factors ,Immunotherapy - Abstract
Biliary tract cancers (BTCs) are a heterogenous group of cancers arising from the biliary tract. The hallmark of these cancers is the advanced stage of presentation and a paucity of durable treatment options. Despite the advances in targeted therapy and immunotherapy in solid tumors, systemic cytotoxic chemotherapy has remained the mainstay for cholangiocarcinomas.With advances in the understanding of the tumor microenvironment, genetic features, and inflammatory milieu, have led to the identification of tumor-infiltrating immune cells as indicators of prognosis and response to treatment in BTC. Through an improved comprehension of immunology, immuno-oncology is becoming another pillar of treatment along with traditional radiation, surgery, cytotoxic chemotherapy, and targeted therapies. This article reviews the evidence for immunotherapy use in cholangiocarcinoma, which still being in infancy, and offers promising new novel options for the management of biliary tract cancers.
- Published
- 2022
12. Tumor response-speed heterogeneity as a novel prognostic factor in patients with mCRC
- Author
-
Junjia Liu, Xuefeng Wang, Ibrahim H. Sahin, Iman Imanirad, Seth I. Felder, Richard D. Kim, and Hao Xie
- Abstract
PurposeDifferential tumor response to therapy is partially attributed to tumor heterogeneity. Additional efforts are needed to identify tumor heterogeneity parameters in response to therapy that are easily applicable in clinical practice. We aimed to describe tumor response-speed heterogeneity and evaluate its prognostic value in patients with metastatic colorectal cancer (mCRC).Patients and MethodsIndividual patient data from Amgen (NCT00364013) and Sanofi (NCT00305188; NCT00272051) trials were retrieved from Project Data Sphere. Patients in the Amgen 5-fluorouracil, leucovorin, oxaliplatin (FOLFOX) arm were used to establish response-speed heterogeneity. Its prognostic value was subsequently validated in the Sanofi FOLFOX arms and the Amgen panitumumab + FOLFOX arm. Kaplan-Meier method and Cox proportional hazards models were used for survival analyses.ResultsPatients with high response-speed heterogeneity in the Amgen FOLFOX cohort had significantly shorter (PPPConclusionTumor response-speed heterogeneity to first-line chemotherapy was a novel prognostic factor associated with early disease progression and shorter survival in patients with mCRC.Implications for PracticeRoutine clinical decision making heavily relies on radiographic assessment of disease response to therapy. For patients with heterogeneous tumors, the degree and kinetics of individual tumor response to the same therapy can sometimes be vastly different. We explored a novel quantitative parameter to describe response-speed heterogeneity by utilizing individual patient data from previous clinical trials. This parameter was an independent prognostic factor associated with early disease progression and shorter survival. Complementary to existing molecular and radiographic tumor heterogeneity parameters, it may help practicing oncologists describe tumor response disparity and serve as a new prognostic factor for patients with mCRC.
- Published
- 2022
- Full Text
- View/download PDF
13. A phase 1/2 trial of ibrutinib in combination with pembrolizumab in patients with mismatch repair proficient metastatic colorectal cancer
- Author
-
James Yu, Estrella Carballido, Richard D. Kim, Maria Elena Martinez, Iman Imanirad, Michael J. Schell, Jun-Min Zhou, Elaine Tan, Dae-Won Kim, Rutika Mehta, and Jonathan R. Strosberg
- Subjects
Adult ,Male ,Oncology ,Cancer Research ,medicine.medical_specialty ,Maximum Tolerated Dose ,Colorectal cancer ,medicine.medical_treatment ,Pembrolizumab ,Adenocarcinoma ,Antibodies, Monoclonal, Humanized ,DNA Mismatch Repair ,Article ,Young Adult ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Piperidines ,Refractory ,Cancer immunotherapy ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Humans ,Neoplasm Metastasis ,Adverse effect ,Aged ,business.industry ,Adenine ,Immunotherapy ,Middle Aged ,medicine.disease ,Progression-Free Survival ,Elevated alkaline phosphatase ,Treatment Outcome ,chemistry ,030220 oncology & carcinogenesis ,Ibrutinib ,Female ,medicine.symptom ,Colorectal Neoplasms ,business - Abstract
BACKGROUND: MMR proficient (pMMR) colorectal cancer (CRC) is usually unresponsive to immunotherapy. Recent data suggest that ibrutinib may enhance the anti-tumour activity of anti-PD-1 immunotherapy. In this study, we evaluated the safety and efficacy of ibrutinib plus pembrolizumab in refractory metastatic CRC. METHODS: This was a phase 1/2 study in patients with refractory metastatic pMMR CRC. The primary endpoints for phases 1 and 2 were maximum tolerated dose (MTD) and disease control rate, respectively. The secondary endpoints were safety, progression-free survival (PFS) and overall survival (OS). RESULTS: A total of 40 patients were enrolled. No dose-limiting toxicity was observed, and MTD was not identified. The highest tested dose of ibrutinib, 560 mg once daily, was combined with a fixed dose of pembrolizumab 200 mg every 3 weeks for the phase 2 portion. The most common grade 3/4 treatment-related adverse events were anaemia (21%), fatigue (8%) and elevated alkaline phosphatase (8%). Among 31 evaluable patients, 8 (26%) achieved stable disease, and no objective response was observed. The median PFS and OS were 1.4 and 6.6 months, respectively. CONCLUSION: Ibrutinib 560 mg daily plus pembrolizumab 200 mg every 3 weeks appears to be well tolerated with limited anti-cancer activity in metastatic CRC. CLINICALTRIALS.GOV IDENTIFIER: NCT03332498.
- Published
- 2021
- Full Text
- View/download PDF
14. Pretreatment CT and PET Radiomics Predicting Rectal Cancer Patients in Response to Neoadjuvant Chemoradiotherapy
- Author
-
Julian Sanchez, Anupam Rishi, Geoffrey Zhang, Jessica M. Frakes, Louis B. Harrison, Zhigang Yuan, Sophie Dessureault, Vladimir Feygelman, Kujtim Latifi, Marissa Frazer, Iman Imanirad, Richard D. Kim, Michal R Tomaszewski, Seth Felder, Sarah E. Hoffe, and Eduardo G. Moros
- Subjects
Tumor Regression Grade ,medicine.diagnostic_test ,business.industry ,Colorectal cancer ,pathologic response ,Exploratory analysis ,medicine.disease ,Logistic regression ,PET ,Oncology ,Radiomics ,radiomics ,Positron emission tomography ,Clinical endpoint ,medicine ,Radiology, Nuclear Medicine and imaging ,rectal cancer ,business ,Nuclear medicine ,Research Paper ,CT ,neoadjuvant chemoradiation therapy ,Neoadjuvant chemoradiotherapy - Abstract
Background: The purpose of this study was to characterize pre-treatment non-contrast computed tomography (CT) and 18 F-fluorodeoxyglucose positron emission tomography (PET) based radiomics signatures predictive of pathological response and clinical outcomes in rectal cancer patients treated with neoadjuvant chemoradiotherapy (NACRT). Material and methods: An exploratory analysis was performed using pre-treatment non-contrast CT and PET imaging dataset. The association of tumor regression grade (TRG) and neoadjuvant rectal (NAR) score with pre-treatment CT and PET features was assessed using machine learning algorithms. Three separate predictive models were built for composite features from CT + PET. Results: The patterns of pathological response were TRG 0 (n = 13; 19.7%), 1 (n = 34; 51.5%), 2 (n = 16; 24.2%), and 3 (n = 3; 4.5%). There were 20 (30.3%) patients with low, 22 (33.3%) with intermediate and 24 (36.4%) with high NAR scores. Three separate predictive models were built for composite features from CT + PET and analyzed separately for clinical endpoints. Composite features with α = 0.2 resulted in the best predictive power using logistic regression. For pathological response prediction, the signature resulted in 88.1% accuracy in predicting TRG 0 vs. TRG 1–3; 91% accuracy in predicting TRG 0–1 vs. TRG 2–3. For the surrogate of DFS and OS, it resulted in 67.7% accuracy in predicting low vs. intermediate vs. high NAR scores. Conclusion: The pre-treatment composite radiomics signatures were highly predictive of pathological response in rectal cancer treated with NACRT. A larger cohort is warranted for further validation.
- Published
- 2021
- Full Text
- View/download PDF
15. Phase 2 study of copanlisib in combination with gemcitabine and cisplatin in advanced biliary tract cancers
- Author
-
Rutika Mehta, Barbara A. Centeno, Richard D. Kim, Elaine S Tan, Biwei Cao, Jongphil Kim, and Taymeyah Al-Toubah
- Subjects
Adult ,Male ,Oncology ,Cancer Research ,medicine.medical_specialty ,medicine.medical_treatment ,Phases of clinical research ,Deoxycytidine ,Disease-Free Survival ,Cholangiocarcinoma ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Humans ,PTEN ,030212 general & internal medicine ,Precision Medicine ,Adverse effect ,Aged ,Phosphoinositide-3 Kinase Inhibitors ,Copanlisib ,Aged, 80 and over ,Cisplatin ,Chemotherapy ,biology ,business.industry ,PTEN Phosphohydrolase ,Gallbladder ,High-Throughput Nucleotide Sequencing ,Middle Aged ,Gemcitabine ,Progression-Free Survival ,Biliary Tract Neoplasms ,Pyrimidines ,chemistry ,030220 oncology & carcinogenesis ,Quinazolines ,Absolute neutrophil count ,biology.protein ,Female ,business ,medicine.drug - Abstract
Background Biliary tract cancer (BTC) has a poor prognosis despite treatment with first-line gemcitabine and cisplatin. In BTC, PI3K/AKT pathway activation has been shown to increase resistance to chemotherapy, which may be overcome with PI3K inhibition. This phase 2 study evaluated the safety and efficacy of copanlisib, a PI3K inhibitor, with gemcitabine and cisplatin in advanced BTCs. The role of PTEN expression in outcomes was also explored. Methods Patients with advanced/unresectable BTC received gemcitabine, cisplatin, and copanlisib as their first-line treatment. The primary endpoint was progression-free survival (PFS) at 6 months. Secondary endpoints were the response rate (RR), median overall survival (OS)/PFS, and safety profile. An assessment of PTEN expression by immunohistochemistry was also performed along with molecular profiling. Results Twenty-four patients received at least 1 dose of the study drug. The PFS rate at 6 months was 51%; the median OS was 13.7 months (95% CI, 6.8-18.0 months), and the median PFS was 6.2 months (95% CI, 2.9-10.1 months). Nineteen patients were evaluable for RR: 6 patients achieved a partial response (31.6%), and 11 (57.9%) had stable disease. The most common grade 3/4 adverse events were a decreased neutrophil count (45.83%), anemia (25%), increased lipase (25%), and hypertension (20.8%). Twenty patients had tissue evaluable for the PTEN status. The PFS for low (n = 9) and high PTEN expression (n = 11) was 8.5 and 4.6 months, respectively (P = .19). The median OS for low and high PTEN expression groups was 17.9 and 7.0 months, respectively (P = .19). Conclusions The addition of copanlisib to gemcitabine and cisplatin does not improve PFS at 6 months. However, future studies using PTEN as a potential biomarker should be considered. Lay summary The addition of copanlisib, a PI3K inhibitor, to standard chemotherapy for advanced biliary tract cancers was assessed for efficacy and safety. Twenty-four patients with advanced biliary tract cancer received treatment in this study. There was no difference in survival with the addition of copanlisib in comparison with standard chemotherapy. Copanlisib may be more effective and increase survival in patients with low PTEN expression levels. Further studies are needed to confirm this. No unexpected adverse events occurred.
- Published
- 2020
- Full Text
- View/download PDF
16. The Application of Circulating Tumor DNA in the Screening, Surveillance, and Treatment Monitoring of Colorectal Cancer
- Author
-
Hao Xie and Richard D. Kim
- Subjects
Oncology ,medicine.medical_specialty ,business.industry ,Colorectal cancer ,Precision medicine ,medicine.disease ,Somatic evolution in cancer ,03 medical and health sciences ,0302 clinical medicine ,DNA profiling ,Surgical oncology ,030220 oncology & carcinogenesis ,Internal medicine ,medicine ,030211 gastroenterology & hepatology ,Surgery ,Epigenetics ,Liquid biopsy ,business ,Treatment monitoring - Abstract
Precision medicine with genetic profiling of tumor tissue has become an essential part of routine clinical practice in colorectal cancer. However, tissue genetic profiling suffers from clonal evolution, tumor heterogeneity, and time needed to deliver critical information for prompt clinical decision making. In contrast, liquid biopsy with plasma circulating tumor DNA provides genetic and epigenetic information from both the primary and metastatic colorectal cancer, which can potentially capture tumor heterogeneity and evolution with time and treatment. In addition, liquid biopsy with circulating tumor DNA is minimally invasive, quicker, and easily repeatable with high patient compliance to provide both qualitative and quantitative molecular information in real-time. We provide an overview on the potential clinical applications of circulating tumor DNA in the screening, surveillance, and treatment monitoring of colorectal cancer.
- Published
- 2020
- Full Text
- View/download PDF
17. Phase II Study of Ensituximab, a Novel Chimeric Monoclonal Antibody, in Adults with Unresectable, Metastatic Colorectal Cancer
- Author
-
Massimo Fantini, Amit Mahipal, Michael A. Morse, Kwong Y. Tsang, Anjum Zaki, B. T. Benjamin Tan, Richard D. Kim, Philip M. Arlen, Jose R. Torrealba, Nilofer S. Azad, Sharon Mavroukakis, Elizabeth Poplin, and Muhammad Shaalan Beg
- Subjects
Adult ,Male ,0301 basic medicine ,Cancer Research ,medicine.medical_specialty ,Nausea ,Anemia ,Colorectal cancer ,medicine.medical_treatment ,Phases of clinical research ,Mucin 5AC ,Gastroenterology ,03 medical and health sciences ,Antineoplastic Agents, Immunological ,0302 clinical medicine ,Refractory ,Internal medicine ,medicine ,Humans ,Infusions, Intravenous ,Adverse effect ,Aged ,Neoplasm Staging ,Chemotherapy ,business.industry ,Antibodies, Monoclonal ,Middle Aged ,medicine.disease ,Progression-Free Survival ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Vomiting ,Female ,medicine.symptom ,Colorectal Neoplasms ,business - Abstract
Purpose: Patients with metastatic colorectal cancer refractory to chemotherapy have limited treatment options. Ensituximab (NEO-102) is a novel chimeric mAb targeting a variant of MUC5AC with specificity to colorectal cancer. Patients and Methods: Single-arm, phase II trial assessed the efficacy and safety of ensituximab in patients with advanced, refractory cancer who expressed MUC5AC antigen in tumor tissue. Ensituximab was administered intravenously every 2 weeks with 3 mg/kg as recommended phase II dose (RP2D). A minimum sample size of 43 patients was required on the basis of the assumption that ensituximab would improve median overall survival (OS) by 7 months using a one-sided significance level of 10% and 80% power. Written informed consent was obtained from all patients. Results: Sixty-three patients with advanced, refractory colorectal cancer were enrolled and 53 subjects were treated in phase II arm. Median age was 58 years and 46% of the patients were female. Among 57 evaluable patients, median OS was 6.8 months. No responses were observed, and stable disease was achieved in 21% of the patients. The most common treatment-related adverse events (AE) at RP2D included fatigue (38%), anemia (30%), nausea (15%), vomiting (11%), increased bilirubin (9%), constipation (8%), decreased appetite (6%), and diarrhea (6%). Serious AEs at least possibly related to ensituximab occurred in 4 patients and included anemia, nausea, increased bilirubin, and hypoxia. No patients discontinued treatment due to drug-related AEs. Conclusions: Ensituximab was well tolerated and demonstrated modest antitumor activity in patients with heavily pretreated refractory colorectal cancer.
- Published
- 2020
- Full Text
- View/download PDF
18. A multi‐institutional phase 2 trial of regorafenib in refractory advanced biliary tract cancer
- Author
-
Dae-Won Kim, Yingmiao Liu, Hanna K. Sanoff, Heloisa P. Soares, Andrew B. Nixon, Andrew Poklepovic, Jongphil Kim, Richard D. Kim, and Jing Lyu
- Subjects
Adult ,Male ,Cancer Research ,medicine.medical_specialty ,Drug-Related Side Effects and Adverse Reactions ,Pyridines ,Population ,Vascular Endothelial Growth Factor D ,Phases of clinical research ,Angiogenesis Inhibitors ,Gastroenterology ,Article ,Metastasis ,Cholangiocarcinoma ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Refractory ,Internal medicine ,Regorafenib ,Clinical endpoint ,medicine ,Humans ,030212 general & internal medicine ,Adverse effect ,education ,Aged ,Aged, 80 and over ,education.field_of_study ,business.industry ,Phenylurea Compounds ,Cancer ,Middle Aged ,medicine.disease ,Progression-Free Survival ,Gene Expression Regulation, Neoplastic ,Biliary Tract Neoplasms ,Oncology ,chemistry ,030220 oncology & carcinogenesis ,Female ,business - Abstract
Background Regorafenib is an oral multikinase inhibitor targeting angiogenesis, oncogenesis, and cancer proliferation/metastasis. This study evaluated the efficacy of regorafenib in refractory biliary tract cancer (BTC) in a multi-institutional phase 2 study. Methods Patients with BTC who progressed on at least 1 line of systemic therapy received regorafenib at 160 mg daily for 21 days on and 7 days off. The primary endpoint was 6-month overall survival (OS), and the secondary endpoints were median OS, progression-free survival (PFS), and objective response rates. Pretreatment plasma was collected for cytokine evaluation. Results A total of 39 patients were enrolled, and 33 were evaluable for efficacy. The median PFS and OS were 3.7 and 5.4 months, respectively, with survival rates of 46.2% at 6 months, 35.9% at 12 months, and 25.6% at 18 months for the intention-to-treat population. For the 33 evaluable patients who received regorafenib for at least 3 weeks, the median PFS and OS were 3.9 and 6.7 months, respectively, with survival rates of 51.5% at 6 months, 39.4% at 12 months, and 27.3% at 18 months. The objective response rate was 9.1%, and the disease control rate was 63.6%. Twenty-eight patients (71.8%) experienced grade 3/4 adverse events. Among the 23 cytokines analyzed, elevated baseline vascular endothelial growth factor D (VEGF-D) was associated with shorter PFS, whereas elevated baseline interleukin 6 (IL-6) and glycoprotein 130 (GP130) were associated with shorter OS. Conclusions Regorafenib demonstrated modest clinical efficacy in heavily pretreated patients with BTC. Further exploration of biomarkers is warranted to identify a group of patients with BTC who may benefit from regorafenib.
- Published
- 2020
- Full Text
- View/download PDF
19. Randomised phase II trial (SWOG S1310) of single agent MEK inhibitor trametinib Versus 5-fluorouracil or capecitabine in refractory advanced biliary cancer
- Author
-
Milind Javle, Vaibhav Sahai, Edward J. Kim, Abby B. Siegel, Katherine A. Guthrie, Stacey M. Stein, Howard S. Hochster, Ari David Baron, Shannon McDonough, Afsaneh Barzi, Tanios Bekaii-Saab, Anthony B. El-Khoueiry, George P. Keogh, and Richard D. Kim
- Subjects
Male ,0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,Pyridones ,5-Fluorouracil ,medicine.medical_treatment ,Oncology and Carcinogenesis ,Pyrimidinones ,Advanced biliary cancer ,Article ,Capecitabine ,03 medical and health sciences ,0302 clinical medicine ,Clinical Research ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,Humans ,Medicine ,Oncology & Carcinogenesis ,Progression-free survival ,Cancer ,Trametinib ,MEK inhibitor ,Chemotherapy ,business.industry ,Evaluation of treatments and therapeutic interventions ,Middle Aged ,Interim analysis ,Gemcitabine ,Biliary Tract Neoplasms ,030104 developmental biology ,Fluorouracil ,6.1 Pharmaceuticals ,030220 oncology & carcinogenesis ,Public Health and Health Services ,Female ,Digestive Diseases ,business ,medicine.drug - Abstract
Background The rationale for the evaluation of trametinib in advanced biliary cancer (BC) is based on the presence of mitogen-activated protein kinase alterations and on earlier promising results with MEK inhibitors in BC. Methods Patients with histologically proven BC who progressed on gemcitabine/platinum were randomised to trametinib daily (arm 1) versus fluoropyrimidine therapy (infusional 5-fluorouracil or oral capecitabine, arm 2). The primary end-point was overall survival (OS). Secondary end-points included progression free survival (PFS) and response rate. A planned interim futility analysis of objective response was performed on the first 14 patients registered to the trametinib arm. Results The study was stopped early based on the lack of measurable response in the trametinib arm. A total of 44 eligible patients were randomised (24 patients in arm 1 and 20 patients in arm 2). Median age was 62 years and the primary sites of tumour were cholangiocarcinoma (68%) and gallbladder (32%). The overall response rate was 8% (95% CI 0%–19%) in arm 1 versus 10% (95% CI 0%–23%) in arm 2 (p > .99) Median OS was 4.3 months for arm 1 and 6.6 months for arm 2. The median PFS was 1.4 months for arm 1 and 3.3 months for arm 2. Conclusions This is the first prospective randomised study of a targeted agent versus chemotherapy for the second-line treatment of BC. In this unselected population, the interim analysis result of unlikely benefit with trametinib resulted in early closure.
- Published
- 2020
- Full Text
- View/download PDF
20. The Impact of Ramucirumab Treatment on Survival and Quality of Life in Patients with Gastric Cancer
- Author
-
Rutika Mehta, Anuhya Kommalapati, and Richard D. Kim
- Subjects
0301 basic medicine ,Oncology ,Chemotherapy ,medicine.medical_specialty ,business.industry ,medicine.medical_treatment ,Cancer ,medicine.disease ,Dysphagia ,Systemic therapy ,Ramucirumab ,03 medical and health sciences ,chemistry.chemical_compound ,030104 developmental biology ,0302 clinical medicine ,Paclitaxel ,chemistry ,Quality of life ,030220 oncology & carcinogenesis ,Internal medicine ,medicine ,Adenocarcinoma ,medicine.symptom ,business - Abstract
Gastric cancer is the sixth most common cancer and is known to be the fifth-leading cause of cancer-related deaths globally in 2018. Systemic therapy remains the only curative option in advanced gastric carcinoma with the primary goal of improving the Health-related Quality of Life (HRQoL) (including palliation of symptoms such as dysphagia) and prolonging overall survival. Recently, ramucirumab is approved by the United States Food and Drug Administration (US-FDA) as a second-line agent either as monotherapy or in combination with paclitaxel in advanced or metastatic gastric and gastro-esophageal junction adenocarcinoma patients who have progressed on prior treatment with fluoropyrimidine or platinum containing chemotherapy. HRQoL is a subjective term that typically constitutes four components - psychological, social, occupational and physical well being. This has been evaluated as secondary endpoint in the pivotal Phase III trials with ramucirumab. HRQoL measurement can potentially provide additional information for clinical decision making beyond that of traditional medical outcomes. The present work is primarily focused on discussing HRQoL in gastric cancer patients and the impact of ramucirumab on the HRQoL in the patients with advanced gastric cancer. We also summarized the studies that evaluated the benefits of systemic therapies on HRQoL in advanced gastric cancer.
- Published
- 2020
- Full Text
- View/download PDF
21. A phase I/Ib study of regorafenib and nivolumab in mismatch repair proficient advanced refractory colorectal cancer
- Author
-
Richard D. Kim, Bence P. Kovari, Maria Martinez, Hao Xie, Ibrahim H. Sahin, Rutika Mehta, Jonathan Strosberg, Iman Imanirad, Masoumeh Ghayouri, Young-chul Kim, and Dae Won Kim
- Subjects
Cancer Research ,Nivolumab ,Oncology ,Pyridines ,Phenylurea Compounds ,Antineoplastic Combined Chemotherapy Protocols ,Colonic Neoplasms ,Humans ,Exanthema ,Colorectal Neoplasms ,DNA Mismatch Repair - Abstract
In contrast to mismatch repair deficient (dMMR) colorectal cancer (CRC), mismatch repair proficient (pMMR) CRC is usually unresponsive to anti-PD-1 immunotherapy. Recent preclinical data suggest that regorafenib may enhance the antitumor activity of anti-PD-1 immunotherapy. However, the safety and efficacy of regorafenib plus nivolumab have not been established in patients with refractory metastatic pMMR CRC. This study aimed to evaluate the safety and efficacy of regorafenib plus nivolumab in metastatic pMMR metastatic CRC.This was a phase I/Ib study with standard 3 + 3 design plus dose expansion of the maximum tolerated dose (MTD) in patients with refractory metastatic pMMR CRC. Patients were treated with regorafenib combined with nivolumab. The primary end-points were dose-limiting toxicity (DLT) and MTD. The secondary end-points were objective response rate, safety and overall survival (OS).A total of 52 patients were enrolled, and 51 patients received at least one dose of treatment. Three patients experienced DLT (all grade 3 rash). MTD was regorafenib 80 mg and nivolumab 240 mg every 2 weeks. Most common grade 3/4 treatment-related adverse events were hypertension (16%), rash (10%) and anaemia (6%). Among 40 evaluable patients, four (10%) achieved partial response, including one unconfirmed response, 21 (53%) achieved stable disease, and disease control rate was 63%. The median progression-free survival and OS were 4.3 and 11.1 months, respectively.Regorafenib plus nivolumab appears to be well tolerated with limited anticancer activity in metastatic pMMR CRC.ClinicalTrials.gov identifier: NCT03712943.
- Published
- 2022
22. Biomarker analysis from a phase I/I1b study of regorafenib and nivolumab (rego/nivo) in microsatellite stable (MSS) colorectal cancer (CRC)
- Author
-
James Yu, Youngchul Kim, Rutika Mehta, Ruoyu Miao, Jonathan R. Strosberg, Iman Imanirad, Dae Won Kim, and Richard D. Kim
- Subjects
Cancer Research ,Oncology - Abstract
188 Background: Our previous phase I/Ib study revealed modest clinical efficacy of rego/nivo in refractory MSS-CRC, implying benefits in selected populations. This has prompted us to investigate predictive biomarkers. Methods: Pre-treatment tumor samples from the previous phase Ib rego/nivo study were obtained and assessed for mRNA sequencing (RNAseq). Response-associated transcripts were identified using DESeq2 method and annotated using gene ontology (GO) and Kyoto encyclopedia of genes and genomes (KEGG) enrichment analysis. Results: A total of 19 pretreatment tumor samples were analyzed including 14 patients (pts) with disease control (DC) (2 partial response and 12 stable disease) and 5 pts with progression disease (PD). We observed significant upregulation of 89 genes including SFTPB ( P
- Published
- 2023
- Full Text
- View/download PDF
23. A phase II study of atezolizumab (ATEZO) and bevacizumab (BEV) in combination with Y90 TARE in patients (pts) with hepatocellular carcinoma (HCC): Y90+/- BEAT
- Author
-
Renuka V. Iyer, Michael Petroziello, Nainesh Parikh, Richard D. Kim, Thatcher Ross Heumann, Daniel Brown, Kevin Kim, Yixing Jiang, Suvranu Ganguli, Eric Marks, Beau Toskich, Umair Majeed, Shamar Young, Rachna T. Shroff, Moh'd M. Khushman, Andrew Gunn, Filip Banovac, and Aiwu Ruth He
- Subjects
Cancer Research ,Oncology - Abstract
TPS629 Background: The anti–PD-L1 antibody ATEZO prevents PD-L1 from interacting with PD-1 and B7.1, thus reinvigorating antitumor T cell activity. Anti-VEGF BEV can increase dendritic cell maturation, enhance T cell infiltration, and reduce myeloid-derived suppressor cells and regulatory T-cells in tumors. This combination has been FDA approved for first-line treatment of advanced HCC based on the IMbrave150 study. On the other hand, locoregional radiotherapy (e.g., Y90 TARE) enhances the diversity of the intratumoral T cell receptor repertoire. It is the standard of care for pts with intermediate-stage HCC (iHCC). Based on preclinical and clinical data, we hypothesize that the combination of Y90 TARE, BEV, and ATEZO induces synergistic tumor-killing. Methods: This is an open-label, multicenter, randomized phase II study of Y90 TARE and BEV plus ATEZO compared with Y90 TARE alone in pts with unresectable IHCC (NCT04541173). The primary study objective is to assess and compare the progression-free survival (PFS) (per mRECIST 1.1) of pts in each arm. The main secondary objective is to determine the safety and tolerability (CTCAE v5) of TARE combined with ATEZO and BEV in pts with HCC. Exploratory objectives are to assess the role of the immunoscore and PD-L1 expression levels in the prediction of improved clinical outcome in pts receiving Y-90 TARE and BEV plus ATEZO; the composition of tumor-infiltrating immune cell subtypes in predicting response to a chosen therapy; how Y90 therapy affects the proportion of antigen-presenting cells in the tumor; and symptoms experienced by pts receiving TARE and BEV plus ATEZO treatment, using patient-reported outcomes. Eligible pts have either HCC that is not amenable to surgical resection, confirmed by pathology review, or at least BCLC stage B HCC outside of downstaging criteria. Other standard eligibility criteria apply. Pts must have a pretreatment liver biopsy taken and then be randomized 1:1 to TARE (Arm A) or TARE followed by ATEZO and BEV (Arm B). Pts will have TARE mapping during week (wk) 1 and TARE treatment during wk 2. In Arm B, pts will start BEV plus ATEZO 4 wks (±1 wk) after TARE treatment. Pts will have abdominal MRI or CT scans every 12 weeks, CT scans of the chest every 24 wks. Disease progression will be captured by both RECIST 1.1 and mRECIST. We plan to assess the safety of TARE with BEV and ATEZO in the first 10 pts randomized to Arm B for two cycles. If there are no Grade ≥ 3 unexpected toxicities possibly, probably or definitely related to combined TARE plus BEV plus ATEZO, the study will continue to accrue 128 pts in total. Pts will continue study treatment (Arm B) for a total of 24 months from initiation of TARE or until intolerable toxicity or disease progression occur, whichever is earlier. Clinical trial information: NCT04541173 .
- Published
- 2023
- Full Text
- View/download PDF
24. Biomarker analysis to predict response in patients with metastatic mismatch repair proficient colorectal cancer treated with regorafenib and nivolumab
- Author
-
Ruoyu Miao, Dae Won Kim, James Yu, Bence Kovari, Rutika Mehta, Jonathan R. Strosberg, Iman Imanirad, Seema Iyer, Mark Uhlik, Laura E. Benjamin, and Richard D. Kim
- Subjects
Cancer Research ,Oncology - Abstract
228 Background: We previously conducted a phase I/Ib study with regorafenib and nivolumab in patients with refractory metastatic mismatch repair proficient (pMMR) colorectal cancer (CRC). This study aimed to investigate the biomarkers that predict the treatment response. Methods: Out of the 51 patients who received regorafenib and nivolumab, 22 archival pretreatment tumor samples were subjected to the Xerna TME Panel, a machine learning-based RNA-sequencing biomarker assay and were classified into one of four TME biomarker subtypes: Angiogenesis (A), Immune Active (IA), Immune Desert (ID), or Immune Suppressed (IS). Potential predictive biomarkers including the TME subtypes, KRAS (wild type vs mutant), PD-L1 (negative vs. positive, samples with > 1% tumor cells for PD-L1 were considered positive), CD8 expression (low vs. high), and Treg cells (low vs. high) in tumor microenvironment were evaluated for correlation with overall survival (OS), progression free survival (PFS) and disease control rate (DCR, defined as complete response + partial response + stable disease). Results: Among the 22 patients, 16 (72.7%) had liver metastasis and 15 (68.2%) had lung metastasis. KRAS mutation was found in 16 (68.2%) patients. 11/21 (52.4%) were positive for PD-L1. 12 (54.5%) had high CD8 expression, whereas 9/21 (42.9%) had high Treg cells in tumor microenvironment. Ten (45.5%) patients were classified as biomarker-positive (IA + IS subtypes) and 12 (54.5%) were biomarker-negative (A + ID) based on Xerna TME panel. Two (9.1%) patients achieved partial response, 12 (54.5%) had stable disease, and five (22.7%) developed progressive disease. The median PFS was 5.6 months and median OS was 13.1 months. No significant correlation was observed between RAS mutation (p = 0.664, p = 0.609), PD-L1 expression (p = 0.287, p = 0.173), CD8 (p = 0.152, p = 0.456) and PFS or OS. Low Treg was found to be associated with prolonged PFS (median: 9.8 vs. 1.9 months, p = 0.011) but not OS (p = 0.280). Similarly, only low Treg level was related with DCR (83.3% vs. 33.3%, p = 0.032). While not reaching statistical significance, Xerna TME biomarker-positive patients showed trends for higher median PFS (7.9 months vs. 4.1 months, p = 0.254), median OS (15.75 months vs. 11.9 months, p = 0.378), and higher DCR (70% vs. 58%, p = 0.675) compared to biomarker-negative patients. Additionally, the two patients with partial responses were Xerna TME biomarker-positive. Conclusions: Our study demonstrated that low Treg in tumor microenvironment is correlated with better prognosis in patients with refractory metastatic pMMR CRC who were treated with regorafenib plus nivolumab. Xerna TME panel analysis of these patients also showed trends for predictive clinical benefit. Prospective and larger cohort studies are needed to better define predictive biomarkers for this combination in the future.
- Published
- 2023
- Full Text
- View/download PDF
25. The role of socioeconomic deprivation in gastrointestinal cancer clinical trial enrollment at an NCI-designated comprehensive cancer center
- Author
-
Daniel Strebig, Taymeyah E. Al-Toubah, Emily Coughlin, Rahul Mhaskar, Sylea Lowery, Ebin Mathew, Mitchell Capelli, Aishwarya Pattnaik, Kea Turner, Kedar Kirtane, Amina Dhahri, Richard D. Kim, Jennifer B. Permuth, Susan Thomas Vadaparampil, Jason B. Fleming, Jonathan R. Strosberg, and Benjamin Daniel Powers
- Subjects
Cancer Research ,Oncology - Abstract
785 Background: Socioeconomic deprivation has been described as a barrier to cancer clinical trial participation. However, few studies have examined socioeconomic deprivation using patient-level or granular geocoded designations. To overcome this challenge, the Area Deprivation Index (ADI) was used to assess neighborhood socioeconomic deprivation in a cohort of gastrointestinal cancer clinical trial patients at an NCI-Designated Comprehensive Cancer Center. Methods: Patients enrolled in a gastrointestinal cancer clinical trial from 2008 to 2019 with an identifiable ADI national rank were identified. Socioeconomic deprivation was assessed using the ADI, a publicly available, validated dataset that ranks census block groups into percentiles using variables such as income, education, employment, and housing characteristics. For this study, ADI was categorized as quintiles listed in the table. Statistical analyses included Chi-square and Kruskal-Wallis tests. Results: The median age of the cohort (N=1,334) was 62.0 years. Most patients were male (54.3%). Race included White (88.2%), African American (6.8%), and Asian (1.5%) patients. Hispanic/Latinx patients made up 6.7% of the cohort. The median ADI was 46. The proportion of enrollees from lowest to highest ADI quintile was 11.2%, 29.3%, 27.3%, 19.1%, and 13.1%. Trial enrollment differed by ADI and age (p=0.019), gender (p=0.042), race (p
- Published
- 2023
- Full Text
- View/download PDF
26. Effect of immunotherapy on the survival outcomes in tumor mutational burden-high (TMB-H) microsatellite stable (MSS) metastatic colorectal cancer (mCRC): A single-institution experience
- Author
-
Ola Gaber, Canan Karan, Christine Marie Walko, Todd C Knepper, Richard D. Kim, and Ibrahim Halil Sahin
- Subjects
Cancer Research ,Oncology - Abstract
239 Background: The benefit of immunotherapy for patients with mCRC with high tumor mutational burden (TMB-H) has been widely debated. In 2020, the FDA approved the use of pembrolizumab for the treatment of patients with TMB-H unresectable or metastatic solid tumors, with TMB-H defined as ≥10 mutations/Mb on a commercial tissue-based assay, based on KEYNOTE 158 results. However, the clinical value of applying this universal cut off to mCRC needs further investigation. Methods: We queried Moffitt Cancer Center (MCC) databases for patients with MSS mCRC, harboring TMB-H (tested with tissue and/or liquid biopsies) who received immunotherapy between January 2018 and December 2021. Patients with incomplete records were excluded. Clinical data were extracted by trained staff from electronic medical records. Objective response rate was measured by using clinical assessment from chart review. Results: We identified 40 patients with TMB-H MSS mCRC 13 of whom received immune checkpoint inhibitor therapy. Female patients represented 31% (n=4) of the 13 treated patients. Median age for the patients was 60 years (range 34-71. Thirty-eight percent (n=5) of the primary tumors originated in the right side. Thirty-one percent (n=4) presented with stage 4 CRC at diagnosis. Histopathology was adenocarcinoma in 92% (n=12) and one had neuroendocrine differentiation. Tumors were well-differentiated 8% (n=1), moderately differentiated 46% (n=6), or poorly differentiated 23% (n=3). Four patients (31%) had POLE/ POLD-1 mutations. The objective response rate (ORR) was 31% (4/13) with responses limited to tumors only with POLE/ POLD-1 mutations (100%, 4/4). The median progression free survival (mPFS) was 3.8 months for the overall cohort and 28.5 months for patients with POLE/POLD-1 mutated tumors. Agent specific analysis showed mPFS for patients treated with pembrolizumab was 3.5 months (mean 5.2; range 0.6-15.7), and 3.4 months (mean 12.1; range 0.5-12.1) for patients treated with nivolumab. Five patients received immunotherapy in combination with regorafenib. The mPFS for these patients was 3.4 months (mean 3.8; range 0.5-8.0). Of five patients with TMB>30 (38%), 4 (80%) had POLE/POLD-1 mutated tumors. One case without POLE/POLD-1 mutations, but with a TMB>30 did not experience any response. Conclusions: Although TMB-H demonstrated therapeutic significance in the KEYNOTE 158 study, the utility of 10 mutations/MB as a universal cutoff warrants additional evaluation. Here we report that a TMB-H cutoff value of ≥ 10 for patients with MSS CRC was not associated with clinically meaningful response to immunotherapy, but patients with MSS CRC with POLE/ POLD-1 mutations may be more likely to benefit from immunotherapy.
- Published
- 2023
- Full Text
- View/download PDF
27. FGFR4 inhibitors for the treatment of hepatocellular carcinoma: a synopsis of therapeutic potential
- Author
-
Hao Xie, Diego M Alem Glison, and Richard D. Kim
- Subjects
Pharmacology ,Fibroblast Growth Factors ,Carcinoma, Hepatocellular ,Cell Line, Tumor ,Liver Neoplasms ,Humans ,Pharmacology (medical) ,Receptor, Fibroblast Growth Factor, Type 4 ,General Medicine ,Signal Transduction - Abstract
The mainstay pharmacological approaches to patients with hepatocellular carcinoma (HCC) are tyrosine kinase inhibitors, antiangiogenic agents, and immune checkpoint inhibitors in combination therapy. Aberrant signaling of fibroblast growth factor 19 (FGF19) and its corresponding receptor, fibroblast growth factor receptor 4 (FGFR4), are a driver of HCC cell growth and survival. However, the clinical potential of agents targeting aberrant FGF19/FGFR4 signaling has not been adequately explored.We evaluate the existing literature on aberrant signaling of FGF19/FGFR4 in HCC and address the recent preclinical and clinical advances of selective FGFR4 inhibitors in the treatment of advanced HCC. Our literature search was performed in September 2021 on clinical trials and ongoing studies published in journals or presented in conferences for cancer research.Preclinical studies show selective FGFR4 inhibitors to be highly potent. These inhibitors also show promise in clinical trials and demonstrate manageable on-target side effects. An emphasis should be placed on the development of predictive biomarkers and on enhancing the understanding of primary and acquired resistance mechanisms. This will inspire rationale combination therapy strategies for testing in future clinical trials.
- Published
- 2021
28. Anatomic patterns of recurrence in biliary tract cancers: does primary tumor site matter?
- Author
-
Andrew J. Sinnamon, Anthony C. Wood, Megan A. Satyadi, Catherine V. Levitt, Olivia Hardy, Mintallah Haider, Richard D. Kim, Daniel A. Anaya, and Jason W. Denbo
- Subjects
Oncology ,Gastroenterology ,Original Article - Abstract
BACKGROUND: Recommendations for postoperative surveillance and adjuvant therapy following curative-intent resection for biliary tract cancers—including intrahepatic and extrahepatic cholangiocarcinoma (IHCCA and EHCCA) and primary gallbladder cancer (GBC)—are uniform across primary tumor site. However, these tumors may have distinct patterns of recurrence. METHODS: A retrospective observational cohort study was performed at a specialty cancer center. Patients undergoing resection of IHCCA, EHCCA, and GBC were identified (2005–2020). Recurrence-free survival (RFS) was estimated using Kaplan-Meier and Cox proportional hazard methods. Anatomic patterns of initial site of recurrence were described and compared. RESULTS: There were 142 patients included; 50 IHCCA, 32 EHCCA, and 60 GBC. Median RFS was 30.8 months, which was not significantly different between IHCCA, EHCCA, or GBC in univariate analysis or after adjustment. Nodal positivity was significantly associated with poor RFS (HR 3.92, P≤0.001). The most common initial site of recurrence overall was intrahepatic (n=49/64, 77%), in isolation (n=32) or synchronous with other site of recurrence (n=17). Significant differences in anatomic pattern of recurrence were observed (P=0.049) with IHCCAs more commonly recurring with simultaneous hepatic-pulmonary disease (n=5/22, 23%; EHCCA n=2/19, 10%; GBC n=1/23, 4%), GBC more commonly recurring within the porta (n=7/23, 30%; IHCCA n=0; EHCCA n=1/19, 5%), and EHCCA more commonly recurring within the peritoneum (n=5/19, 26%; IHCCA n=2/22, 9%, GBC n=2/23, 9%). CONCLUSIONS: Patterns of initial recurrence appear to differ between primary tumor site, likely reflecting underlying differences in anatomy and biology. These data could help inform future studies for adjuvant therapy as well as timing and anatomic focus for surveillance imaging.
- Published
- 2021
29. Acquired On-Target Clinical Resistance Validates FGFR4 as a Driver of Hepatocellular Carcinoma
- Author
-
Oleg Schmidt-Kittler, Michael Sheets, Yoon-Koo Kang, Chandrasekhar V. Miduturu, Richard D. Kim, Andy Boral, Joseph L. Kim, Nooreen Rubin, Neil Bifulco, Emily Rozsahegyi, Cori Ann Sherwin, Megan A. Hatlen, Natasja Brooijmans, Marion Dorsch, Hongliang Shi, Beni B. Wolf, Klaus P. Hoeflich, Christoph Lengauer, and Timothy J. Guzi
- Subjects
0301 basic medicine ,Mutation ,business.industry ,Kinase ,medicine.medical_treatment ,FGF19 ,Fibroblast growth factor receptor 4 ,Drug resistance ,medicine.disease_cause ,medicine.disease ,Targeted therapy ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Oncology ,030220 oncology & carcinogenesis ,Hepatocellular carcinoma ,medicine ,Cancer research ,Carcinoma ,business - Abstract
Hepatocellular carcinoma (HCC) is a leading cause of cancer mortality worldwide with no clinically confirmed oncogenic driver. Although preclinical studies implicate the FGF19 receptor FGFR4 in hepatocarcinogenesis, the dependence of human cancer on FGFR4 has not been demonstrated. Fisogatinib (BLU-554) is a potent and selective inhibitor of FGFR4 and demonstrates clinical benefit and tumor regression in patients with HCC with aberrant FGF19 expression. Mutations were identified in the gatekeeper and hinge-1 residues in the kinase domain of FGFR4 upon disease progression in 2 patients treated with fisogatinib, which were confirmed to mediate resistance in vitro and in vivo. A gatekeeper-agnostic, pan-FGFR inhibitor decreased HCC xenograft growth in the presence of these mutations, demonstrating continued FGF19–FGFR4 pathway dependence. These results validate FGFR4 as an oncogenic driver and warrant further therapeutic targeting of this kinase in the clinic. Significance: Our study is the first to demonstrate on-target FGFR4 kinase domain mutations as a mechanism of acquired clinical resistance to targeted therapy. This further establishes FGF19–FGFR4 pathway activation as an oncogenic driver. These findings support further investigation of fisogatinib in HCC and inform the profile of potential next-generation inhibitors. See related commentary by Subbiah and Pal, p. 1646. This article is highlighted in the In This Issue feature, p. 1631
- Published
- 2019
- Full Text
- View/download PDF
30. First-in-Human Phase I Study of Fisogatinib (BLU-554) Validates Aberrant FGF19 Signaling as a Driver Event in Hepatocellular Carcinoma
- Author
-
Vincenzo Mazzaferro, Jung Hwan Yoon, Chia Jui Yen, Sunil Sharma, Max Sung, Josep M. Llovet, Tim Meyer, Meera Tugnait, Sandrine Faivre, Stephen L. Chan, Antoine Hollebecque, Daniel H. Palmer, Christoph Lengauer, Teresa Macarulla, Margit Hagel, Ho Yeong Lim, Joong-Won Park, Su Pin Choo, Andrew X. Zhu, Lynn G. Feun, Cori Ann Sherwin, Debashis Sarker, Thomas Yau, Melissa Manoogian, Yoon-Koo Kang, Hongliang Shi, Oleg Schmidt-Kittler, Nicolas Stransky, Zhong Zhe Lin, Beni B. Wolf, Nancy E. Kohl, Richard D. Kim, Jean-François Dufour, Joerg Trojan, and Klaus P. Hoeflich
- Subjects
0301 basic medicine ,Oncology ,medicine.medical_specialty ,business.industry ,Nausea ,FGF19 ,medicine.disease ,Clinical trial ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,030220 oncology & carcinogenesis ,Hepatocellular carcinoma ,Internal medicine ,medicine ,Carcinoma ,Vomiting ,Biomarker (medicine) ,medicine.symptom ,business ,Adverse effect - Abstract
Outcomes for patients with advanced hepatocellular carcinoma (HCC) remain poor despite recent progress in drug development. Emerging data implicate FGF19 as a potential HCC driver, suggesting its receptor, FGFR4, as a novel therapeutic target. We evaluated fisogatinib (BLU-554), a highly potent and selective oral FGFR4 inhibitor, in a phase I dose-escalation/dose-expansion study in advanced HCC using FGF19 expression measured by IHC as a biomarker for pathway activation. For dose escalation, 25 patients received 140 to 900 mg fisogatinib once daily; the maximum tolerated dose (600 mg once daily) was expanded in 81 patients. Fisogatinib was well tolerated; most adverse events were manageable, grade 1/2 gastrointestinal events, primarily diarrhea, nausea, and vomiting. Across doses, the overall response rate was 17% in FGF19-positive patients [median duration of response: 5.3 months (95% CI, 3.7–not reached)] and 0% in FGF19-negative patients. These results validate FGFR4 as a targetable driver in FGF19-positive advanced HCC. Significance: Fisogatinib elicited clinical responses in patients with tumor FGF19 overexpression in advanced HCC. These results validate the oncogenic driver role of the FGFR4 pathway in HCC and the use of FGF19 as a biomarker for patient selection. See related commentary by Subbiah and Pal, p. 1646. This article is highlighted in the In This Issue feature, p. 1631
- Published
- 2019
- Full Text
- View/download PDF
31. A phase I pharmacokinetic study of belinostat in patients with advanced cancers and varying degrees of liver dysfunction
- Author
-
Laura K. Fogli, Geraldine O'Sullivan Coyne, William R. Schelman, Richard D. Kim, Nancy Moore, Bassel F. El-Rayes, Chandra P. Belani, Naoko Takebe, Heinz-Josef Lenz, James H. Doroshow, Cindy L. O'Bryant, Sanjay Goel, Jan H. Beumer, Alice P. Chen, Larry Rubinstein, Richard Piekarz, Brian F. Kiesel, Afshin Dowlati, Vincent Chung, Shivaani Kummar, Ulka N. Vaishampayan, and Joseph Tuscano
- Subjects
Male ,drug safety ,Hydroxamic Acids ,030226 pharmacology & pharmacy ,Gastroenterology ,Severity of Illness Index ,Liver disease ,chemistry.chemical_compound ,0302 clinical medicine ,Neoplasms ,Pharmacology (medical) ,030212 general & internal medicine ,Infusions, Intravenous ,Sulfonamides ,Liver Diseases ,Histone deacetylase inhibitor ,Middle Aged ,3. Good health ,Liver ,Toxicity ,Original Article ,Female ,liver disease ,Adult ,medicine.medical_specialty ,Maximum Tolerated Dose ,medicine.drug_class ,Metabolic Clearance Rate ,Drug Administration Schedule ,03 medical and health sciences ,Pharmacokinetics ,Internal medicine ,medicine ,Humans ,Dosing ,histone deacetylase inhibitor ,Aged ,Neoplasm Staging ,Pharmacology ,Dose-Response Relationship, Drug ,business.industry ,Original Articles ,medicine.disease ,drug metabolism ,Histone Deacetylase Inhibitors ,anticancer drugs ,chemistry ,Liver function ,business ,Belinostat ,Drug metabolism - Abstract
Aims The histone deacetylase inhibitor belinostat has activity in various cancers. Because belinostat is metabolized by the liver, reduced hepatic clearance could lead to excessive drug accumulation and increased toxicity. Safety data in patients with liver dysfunction are needed for this drug to reach its full potential in the clinic. Methods We performed a phase 1 trial to determine the safety, maximum tolerated dose (MTD) and pharmacokinetics of belinostat in patients with advanced cancer and varying degrees of liver dysfunction. Results Seventy-two patients were enrolled and divided into cohorts based on liver function. In patients with mild dysfunction, the MTD was the same as the recommended phase 2 dose (1000 mg/m2 /day). Belinostat was well tolerated in patients with moderate and severe liver dysfunction, although the trial was closed before the MTD in these cohorts could be determined. The mean clearance of belinostat was 661 mL/min/m2 in patients with normal liver function, compared to 542, 505 and 444 mL/min/m2 in patients with mild, moderate and severe hepatic dysfunction. Although this trial was not designed to assess clinical activity, of the 47 patients evaluable for response, 13 patients (28%) experienced stable disease. Conclusion While a statistically significant difference in clearance indicates increased belinostat exposure with worsening liver function, no relationship was observed between belinostat exposure and toxicity. An assessment of belinostat metabolites revealed significant differences in metabolic pathway capability in patients with differing levels of liver dysfunction. Further studies are needed to establish formal dosing guidelines in this patient population.
- Published
- 2019
32. The Role of Angiogenesis in Hepatocellular Carcinoma
- Author
-
Weijing Sun, Michelle Mynderse, Michael A. Morse, Richard D. Kim, Paolo Abada, Aiwu Ruth He, and Richard S. Finn
- Subjects
Vascular Endothelial Growth Factor A ,0301 basic medicine ,Cancer Research ,Carcinoma, Hepatocellular ,Angiogenesis ,Vascular Endothelial Growth Factor Receptor ,Angiogenesis Inhibitors ,Neovascularization ,Pathogenesis ,03 medical and health sciences ,0302 clinical medicine ,Carcinoma ,Humans ,Medicine ,Clinical significance ,Angiostatins ,Protein Kinase Inhibitors ,Predictive biomarker ,Neovascularization, Pathologic ,business.industry ,Liver Neoplasms ,Sorafenib ,Prognosis ,medicine.disease ,digestive system diseases ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Hepatocellular carcinoma ,Cancer research ,medicine.symptom ,business - Abstract
Hepatocellular carcinoma (HCC) accounts for about 90% of all primary liver cancers and is the second leading cause of cancer-related deaths worldwide. The hypervascular nature of most HCC tumors underlines the importance of angiogenesis in the pathobiology of these tumors. Several angiogenic pathways have been identified as being dysregulated in HCC, suggesting they may be involved in the development and pathogenesis of HCC. These data provide practical targets for systemic treatments such as those targeting the vascular endothelial growth factor receptor and its ligand. However, the clinical relevance of other more recently identified angiogenic pathways in HCC pathogenesis or treatment remains unclear. Research into molecular profiles and validation of prognostic or predictive biomarkers will be required to identify the patient subsets most likely to experience meaningful benefit from this important class of agents.
- Published
- 2019
- Full Text
- View/download PDF
33. Network meta-analysis of nivolumab plus ipilimumab in the second-line setting for advanced hepatocellular carcinoma
- Author
-
Jinlin Song, Richard D. Kim, Keith A. Betts, Keith D. Huff, Muhan Yuan, Alexander Marshall, Neehar D. Parikh, Jing Zhao, and Aozhou Wu
- Subjects
medicine.medical_specialty ,Carcinoma, Hepatocellular ,Cabozantinib ,Network Meta-Analysis ,Urology ,Ipilimumab ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Regorafenib ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Overall survival ,Humans ,030212 general & internal medicine ,business.industry ,Health Policy ,Hazard ratio ,Liver Neoplasms ,medicine.disease ,Nivolumab ,chemistry ,030220 oncology & carcinogenesis ,Hepatocellular carcinoma ,Meta-analysis ,business ,medicine.drug - Abstract
Aims: To compare the efficacy of nivolumab 1 mg/kg + ipilimumab 3 mg/kg with regorafenib 160 mg, cabozantinib 60 mg and nivolumab 3 mg/kg monotherapy for second-line treatment of advanced hepatocellular carcinoma. Materials & methods: Indirect comparison using network meta-analysis and propensity score weighting. Results: Nivolumab 1 mg/kg + ipilimumab 3 mg/kg had significantly higher objective response rate (median 31.2% [95% credible interval: 19.6–44.5%]) than cabozantinib (4.2% [2.0–6.5%]) and regorafenib (4.8% [1.1–8.3%]), and significantly longer overall survival (cabozantinib: hazard ratio: 0.46 [95% credible interval: 0.27–0.79]; regorafenib: 0.56 [0.32–0.97]). Nivolumab 1 mg/kg + ipilimumab 3 mg/kg had significantly better objective response rate (difference 21.0% [4.5–37.5%]) and overall survival (hazard ratio: 0.58 [0.35–0.96]) than nivolumab monotherapy. Conclusion: Nivolumab 1 mg/kg + ipilimumab 3 mg/kg had a superior efficacy versus cabozantinib 60 mg, regorafenib 160 mg and nivolumab 3 mg/kg monotherapy as second-line therapy for advanced hepatocellular carcinoma.
- Published
- 2021
34. An open-label, multicenter, randomized phase II study of atezolizumab and bevacizumab with Y90 TARE in patients with unresectable hepatocellular carcinoma (HCC)
- Author
-
Aiwu Ruth He, Filip Banovac, Renuka V. Iyer, Michael Petroziello, Daniel Brown, Laura Williams Goff, Richard D. Kim, Nainesh Parikh, Beau Toskich, Kevin Kim, Yixing Jiang, Suvranu Ganguli, Matthew H. Kulke, Samantha Ann Armstrong, Matthew Johnson, Rachna T. Shroff, and Gregory Woodhead
- Subjects
Cancer Research ,Oncology - Abstract
TPS4177 Background: The anti–PD-L1 antibody atezolizumab (ATEZO) prevents PD-L1 from interacting with PD-1 and B7.1, thus reinvigorating antitumor T cell activity. Anti-VEGF bevacizumab (BEV) increases dendritic cell maturation, enhances T cell infiltration, and reduces myeloid-derived suppressor cells and regulatory T cells in tumors. ATEZO + BEV is FDA approved for first-line treatment of advanced HCC based on the IMbrave 150 study. Locoregional radiotherapy (e.g., Y90 TARE) enhances the diversity of the intratumoral T cell receptor repertoire and increases tumor antigen release. We hypothesize that the Y90 TARE + BEV + ATEZO combination induces synergistic tumor killing and prolongs progression-free survival in patients (pts) receiving Y90 TARE (HR = 0.6 when compared to Y-90 TARE alone). Methods: Eligible pts have HCC that cannot be surgically resected (confirmed by pathology review), is at least BCLC stage B, and is outside Milan criteria. Other requirements include ECOG PS of 0-1 at screening, measurable disease by RECIST 1.1, no prior systemic therapy, and FLR estimated at ≥ 40% post local therapy. Pts must have a pretreatment liver biopsy taken and then be randomized 1:1 to TARE (Arm A) or TARE + ATEZO + BEV (Arm B). Pts will have TARE mapping followed by TARE treatment. In Arm B, pts will begin TARE treatment followed by BEV + ATEZO (4 wks [± 1 wk] later). Pts will have abdominal MRI or CT scans every 12 weeks and CT scans of the chest every 24 wks. The primary study objective is to assess and compare pts' progression-free survival (per mRECIST 1.1) in each study arm. The main secondary objective is to determine the safety and tolerability (CTCAE v5) of pts in Arm B. Exploratory objectives are to define the use of cellular and circulating biomarkers in the prediction of improved clinical outcomes of pts in Arm B. Symptoms experienced by pts in both arms using patient-reported outcomes will be assessed. Disease progression will be captured by both RECIST 1.1 and mRECIST. We plan to assess the safety of Y90 TARE + BEV + ATEZO in the first 10 pts randomized to Arm B for two cycles, and if there are no grade ≥ 3 unexpected toxicities possibly, probably, or definitely related to combined TARE + BEV + ATEZO, continue to accrue 128 pts in total (current enrollment n- = 5). Pts will continue study treatment (Arm B) for a total of 24 months from initiation of TARE or until intolerable toxicity or disease progression occur, whichever is earlier. Enrollment began in September 2020. Clinical trial information: NCT04541173.
- Published
- 2022
- Full Text
- View/download PDF
35. Pembrolizumab (pembro) plus mFOLFOX7 or FOLFIRI for metastatic colorectal cancer (CRC) in KEYNOTE-651: Long-term follow-up of cohorts B and D
- Author
-
Richard D. Kim, Mustapha Tehfe, Petr Kavan, Jorge Chaves, Jeremy S. Kortmansky, Eric Xueyu Chen, Christopher Hanyoung Lieu, Lucas Wong, Marwan Fakih, Kristen Renee Spencer, Qing Zhao, Raluca Predoiu, Chenxiang Li, David Carpenter, Pierre Leconte, and E. Gabriela Chiorean
- Subjects
Cancer Research ,Oncology - Abstract
3521 Background: Response to antiPD-1 monotherapy is poor in patients (pts) with advanced microsatellite stable (MSS)/mismatch-repair proficient (pMMR) CRC. Combination of chemotherapy with antiPD-1 pembro may potentiate the antitumor immune response and provide greater antitumor activity than either agent alone. Combination of pembro and mFOLFOX7 or FOLFIRI in the ongoing phase 1b multicohort KEYNOTE-651 (NCT03374254) study in metastatic MSS/pMMR CRC showed antitumor activity. We present results with approximately 20 mo of additional follow-up. Methods: Pts had metastatic MSS/pMMR CRC and were previously untreated (cohort B) or received 1 prior line including a fluoropyrimidine + oxaliplatin (cohort D). Pts received pembro 200 mg Q3W + mFOLFOX7 Q2W (cohort B) or pembro 200 mg Q3W + FOLFIRI Q2W (cohort D). Primary end points were safety (DLT) and RP2D. Secondary end point was ORR per RECIST v1.1 by investigator review. DOR, DCR, and PFS per RECIST v1.1, and OS were exploratory end points. Results: Median study follow-up (range) at data cutoff (Oct 15, 2021) was 30.2 mo (25.0-43.6) for cohort B (n = 31) and 33.5 mo (25.2-43.2) for cohort D (n = 32). Treatment was discontinued in 29 pts (94%) in cohort B and 28 pts (88%) in cohort D, mostly because of PD (61% and 66%, respectively). At prior analysis (Feb 10, 2020), RP2D was confirmed as the starting dose in both cohorts; no new DLT had occurred. In cohort B, gr 3/4 TRAEs occurred in 18 pts (58%), most commonly neutropenia and decreased neutrophil count (both n = 5; 16%); 19 pts (61%) discontinued drug because of a TRAE. In cohort D, gr 3/4 TRAEs occurred in 17 pts (53%), most commonly, neutropenia (n = 7; 22%), diarrhea and fatigue (both n = 4; 13%); 3 pts (9%) discontinued drug because of a TRAE. There were no gr 5 TRAEs in either cohort. Efficacy by cohort and KRAS mutation status are below (Table). PD-L1 data and DNA/RNA-based biomarker data including GEP, consensus signatures, TMB, and LoF mutations will be included in the presentation. Conclusions: After 2.5 y of follow-up, the combination of pembro with either mFOLFOX7 or FOLFIRI continued to demonstrate a manageable safety profile with no new safety signals. Efficacy data from these single-arm cohorts appear comparable to historical data for current SOC. Clinical trial information: NCT03374254. [Table: see text]
- Published
- 2022
- Full Text
- View/download PDF
36. Pembrolizumab (pembro) plus binimetinib (bini) with or without chemotherapy (chemo) for metastatic colorectal cancer (mCRC): Results from KEYNOTE-651 cohorts A, C, and E
- Author
-
Eric Xueyu Chen, Petr Kavan, Mustapha Tehfe, Jeremy S. Kortmansky, Michael B. Sawyer, E. Gabriela Chiorean, Christopher Hanyoung Lieu, Blase N. Polite, Lucas Wong, Marwan Fakih, Kristen Renee Spencer, Jorge Chaves, Chenxiang Li, David Carpenter, Pierre Leconte, and Richard D. Kim
- Subjects
Cancer Research ,Oncology - Abstract
3573 Background: Response to antiPD-1 monotherapy is poor in microsatellite stable (MSS)/mismatch-repair proficient (pMMR) mCRC; the combination of antiPD-1 pembro + anti-MEK bini, with or without chemo, may improve upon this limited response. KEYNOTE-651 (NCT03374254) is an open-label phase 1b multicenter trial of pembro + bini (cohort A) or pembro + bini + chemo (mFOLFOX7 in cohort C, FOLFIRI in cohort E) in MSS/pMMR mCRC. Preliminary results from the dose-finding phase at 2 dose levels (DL) of bini are presented. Methods: Patients (pts) had MSS/pMMR mCRC and must have been previously treated with fluoropyrimidine, irinotecan, and oxaliplatin in cohort A, or with fluoropyrimidine + oxaliplatin-based regimen in cohort E; pts were previously untreated in cohort C. Pts received pembro 200 mg Q3W + bini 30 mg BID (cohort A, DL1), pembro 200 mg Q3W+ bini 30 mg BID + mFOLFOX7 Q2W (cohort C, DL1) or pembro 200 mg Q3W + bini 30 mg BID + FOLFIRI Q2W (cohort E, DL1). Bini dose escalation to 45 mg BID (DL2) was planned in cohorts A, C, and E, with a target dose-limiting toxicity (DLT) of 30%. Primary end point was safety (DLT). Secondary end point was ORR. DCR, PFS, and OS were exploratory. ORR, DCR, and PFS were assessed by investigator per RECIST v1.1. Results: Median study follow-up at data cutoff (Oct 15, 2021) was 36 mo (range, 32-43) for cohort A, 17 mo (2-24) for cohort C, and 11 mo (2-25) for cohort E. In cohort A, 1/6 pts (17%) had DLT at DL1; no DLT occurred in 14 pts (0%) at DL2. In cohort A, gr 3/4 TRAEs occurred in 3/6 pts (50%) at DL1 and 8/14 pts (57%) at DL2. In cohort C, 3/9 evaluable pts (33%) had DLT at DL1; thus, bini dose was not escalated to DL2. In cohort C, gr 3/4 TRAEs occurred in 9/11 total pts (82%). In cohort E, 1/5 evaluable pts (20%) had DLT at DL1 and 5/10 evaluable pts (50%) had DLT at DL2. Enrollment was stopped in cohort E, DL2 and bini dose was de-escalated to DL1; 2/4 additional pts (50%) had DLT at DL1 (total 3/9 pts [33%] had DLT in cohort E, DL1). In cohort E, gr 3/4 TRAEs occurred in 5/9 pts (56%) at DL1 and 10/11 total pts (91%) at DL2. No gr 5 TRAEs occurred in any cohort. ORR was 0% in cohort A; limited efficacy was seen in cohorts C and E (Table). Conclusions: Bini could be safely combined with pembro in cohort A. However, with bini + pembro + chemo, the 45-mg dose of bini was not well tolerated and required dose reduction to 30 mg. Addition of bini to pembro + chemo did not improve efficacy; therefore, enrollment was prematurely closed in cohorts C and E. Efficacy by KRAS mutation status will be shown. Clinical trial information: NCT03374254. [Table: see text]
- Published
- 2022
- Full Text
- View/download PDF
37. Efficacy and safety of NT-I7, long-acting interleukin-7, plus pembrolizumab in patients with advanced solid tumors: Results from the phase 2a study
- Author
-
Aung Naing, Hirva Mamdani, Minal A. Barve, Melissa Lynne Johnson, Daniel Morgensztern, Anthony J. Olszanski, Robert A. Wolff, Shubham Pant, Marya F. Chaney, Tolani Adebanjo, Jean Fan, Richard D. Kim, and Scott Kopetz
- Subjects
Cancer Research ,Oncology - Abstract
2514 Background: Checkpoint inhibitors (CPIs) are usually ineffective in patients (pts) with immune-cold microsatellite stable colorectal cancer (MSS-CRC) or pancreatic cancer (PDAC) and in those who progressed on previously treated with antibodies against PD1 or PD-L1. Here, we report the combination of NT-I7 plus pembrolizumab (pembro) on CPI-naïve MSS-CRC and PDAC cohorts, and patients (pts) with CPI-treated triple-negative breast cancer (TNBC), non-small cell lung cancer (NSCLC), and small cell lung cancer (SCLC) cohorts of this ongoing phase 2a trial. Methods: Pts with CPI-naïve relapsed/refractory (R/R) MSS-CRC and PDAC, and CPI-treated R/R TNBC, NSCLC, and SCLC, were enrolled. NT-I7 (efineptakin alfa) 1200 µg/kg intramuscularly every 6 weeks and 200 mg pembro intravenously every 3 weeks were administered until disease progression/unacceptable toxicity. The primary endpoint of the Phase 2a is the objective response rate (ORR), assessed by RECIST v1.1 and iRECIST. The secondary endpoints are duration of response (DoR), disease control rate, progression-free survival, and overall survival. Results: As of 14 Jan 2022, 92 pts with metastatic or locally advanced cancer who had received a median of 3 prior treatments were enrolled in the study; 32 in PDAC, 28 in MSS-CRC, 22 in NSCLC, 6 in TNBC, and 4 in SCLC. The median age was 62 years [29-81], ECOG PS 0 in 23 (25%), 1 in 68 (74%) and 2 in 1 (1%). Among 71 evaluable pts, the median follow up (months) was 7.7, 5.3, 5.0, 3.7, and 2.4 in TNBC, MSS-CRC, NSCLC, PDAC and SCLC, respectively. The ORR was 50% (1/2) in SCLC, 12% (3/25) in MSS CRC, 8% (2/26) in PDAC 6% (1/16) in NSCLC and 0% (0/2) in TNBC per iRECIST; and 50% (1/2) in SCLC, 4% (1/25) in MSS CRC, and 4% (1/26) in PDAC per RECIST 1.1. All 7 responders are ongoing. The two PDAC pts had DoR over 1.35 months (mos) and 6.64 mos with the best tumor reduction 100% and 72% respectively. The one SCLC pt had DoR over 1.5 mos with the tumor reduction 67%. The one NSCLC pt had DoR over 2.73 mos with the tumor reduction 60%, and the three MSS-CRC pts had DoR 6.34, 2.96, and 0.03 mos with the tumor reduction 60%, 56%, and 43% respectively. A sustained and significant (approximate 3X from baseline) increase of peripheral lymphocytes in all arms was observed as shown in our previous report. Among 92 treated pts, NT-I7-related adverse events (AEs) occurred in 67 (72.8%) pts, including 52 (56.5%) Grade (G)1-2, 13 (14.1%) G3, and 2 (2.2%) G4. There were no NT-I7 related G5 AEs. Additional updated efficacy, safety and biomarker data will be presented. Conclusions: The combination of NT-I7 and pembro demonstrated antitumor activity and manageable toxicity profile in heavily pretreated pts with CPI-naïve MSS-CRC and PDAC and CPI-treated TNBC, NSCLC, and SCLC, suggesting that the addition of NT-I7 to CPI can overcome the primary resistance to CPI in the former group and acquired resistance in the latter. Clinical trial information: NCT04332653.
- Published
- 2022
- Full Text
- View/download PDF
38. Characterization of NTRK alterations in metastatic colorectal cancer
- Author
-
Canan Karan, Elaine Tan, Humaira Sarfraz, Christine Marie Walko, Richard D. Kim, Todd C Knepper, and Ibrahim Halil Sahin
- Subjects
Cancer Research ,Oncology - Abstract
e15569 Background: Increased molecular profiling ability has resulted in the recognition of important actionable genes that can be used to direct targeted therapies in colorectal cancers (CRC). Neurotrophin receptor tyrosine kinase (NTRK) gene fusions are one of these molecular alterations that are potentially actionable with novel targeted therapeutics. However, other biologically pathogenic NTRK alterations, including point mutations, are not yet actionable in CRC. This study aimed to characterize NTRK alterations, including fusions in metastatic CRC patients. Methods: Molecular characteristics of 917 patients with CRC were collected from the Moffitt Cancer Center Clinical Genomics Action Committee database. Demographic, clinicopathological, and molecular data and treatment history were abstracted from electronic medical records. Mutations with potential oncogenic activity are considered “potentially pathogenic”, while alterations with proven oncogenic activity such as fusions are considered “pathogenic”. Results: There were 917 patients with CRC from November 2013 to December 2021, and 77 of them had NTRK alterations excluding synonymous mutations (8.3%, 77/917). Six patients had potentially pathogenic NTRK alterations, including one NTRK rearrangement (0.6%, 7/917); however, only 1 patient had an actionable pathogenic NTRK1 fusion (0.1%, 1/917), while the others (70) were a variant of unknown significance (VUS). The majority of NTRK alterations were missense mutations (92%; 71/77). The NTRK1 fusion partner was LMNA and occurred in the setting of an MSI-H tumor (3.7%; 1/27). Among patients with pathogenic and potentially pathogenic alterations (N = 7), there was only 1(14%) patient who had low tumor mutation burden (TMB) (< 10 mut/MB). Most patients were older than 50 years (70.1%, 54/77) and male (58.4%, 45/77). Thirty-six patients (46..7%) had right-sided tumor, while 41 patients (53.2%) left-sided. Across all NTRK alterations cohort, MSI-H was found in 10.3% (7/77), and POLE/POLD1 pathogenic mutations were seen in 6.4% (5/77) patients. Conclusions: NTRK gene fusion is a relatively rare event (< 1%) in CRC, including the MSI-H subtype. The majority of NTRK alterations in CRC are VUS, and they are not actionable, and they tend to be seen in the tumors with high TMB (TMB > 10). Hypermutator tumors cause frequent VUS alterations in the NTRK gene with unknown clinical relevance.
- Published
- 2022
- Full Text
- View/download PDF
39. Clinical and molecular characterization of fusion genes in colorectal cancer
- Author
-
Canan Karan, Elaine Tan, Humaira Sarfraz, Christine Marie Walko, Richard D. Kim, Todd C Knepper, and Ibrahim Halil Sahin
- Subjects
Cancer Research ,Oncology - Abstract
e15568 Background: Next-generation sequencing (NGS) based molecular profiling technologies have revealed several oncogenic fusion genes that are actionable with small molecule inhibitors leading to practice change, particularly in lung cancer. The molecular and clinical characteristics of these gene fusions are not well defined in colorectal cancer patients (CRC). In this study, we aimed to define clinical and molecular characteristics of fusion genes in patients with CRC who underwent molecular profiling. Methods: Molecular characteristics of tissue confirmed 917 CRC patients were retrieved from the Moffit Cancer Center Clinical Genomics Action Committee database. Patients’ demographic and clinicopathological features and treatment history were collected from the database. All fusion genes were shown by hybridization-based NGS computational algorithms that determined cancer‐related genes, including single‐nucleotide variations, indels, microsatellite instability (MSI) status. Results: Among a total of 917 patients, 24 patients with CRC (2.6%) were found to have at least one fusion gene with a total number of 26 pathogenic fusions. The gene fusions are shown in Table. The most common, potentially targetable, fusion genes in our cohort were (1) RET fusions 0.5% (5/917), (2) ALK fusions 0.4% (4/917), (3) ROS1 fusions 0.2% (2/917), (4) NTRK1 fusion 0.1% (1/917), (5) NRG1 fusion 0.1% (1/917). Fusion genes were more common in MSI-H CRC (N = 27), and 3 (11.1%) patients with MSI-H CRC were found to have fusion genes [(RET (2) and NTRK(1)]. Fusion genes were present in both RAS wild-type (54%; 13/24) and RAS mutant (46%; 11/24) tumors. Most patients were older than 50 years (75%, 18/24) and had left-sided tumor (61.1%) tumor. Conclusions: Fusion genes are rare events in CRC. While fusion genes seem to be more prevalent in MSI-H CRC, RAS status does not correlate with the frequency of fusion genes. Actionable RET and ALK/ROS gene fusion are more common than NTRK fusion genes in this cohort of CRC patients.[Table: see text]
- Published
- 2022
- Full Text
- View/download PDF
40. Disparity of treatment-related adverse events and outcome in patients with early-onset metastatic colorectal cancer (mCRC)
- Author
-
Lingbin Meng, Ram Thapa, Richard D. Kim, Damian A. Laber, and Hao Xie
- Subjects
Cancer Research ,Oncology - Abstract
6560 Background: While the incidence of newly diagnosed early-onset mCRC has been increasing, disparity of treatment-related adverse events (AE) and outcomes of this patient population has been inadequately studied with inconclusive findings. We aimed to evaluate such age-related disparity and explore potential underlying causes. Methods: We used individual patient data from 3 clinical trials in Project Data Sphere where 756 and 467 patients with mCRC received first-line FOLFOX in study 1 (NCT 00305188, NCT00272051) and study 2 (NCT00364013), respectively. Clinical NGS data of 763 patients with mCRC from prospectively maintained Moffitt Clinical Genomics Database were used to assess genomic alterations. Patients were categorized into 3 age groups: 65 years. Continuous and categorical variables were compared with t test and χ2 test, respectively. Kaplan-Meier method and log-rank test were used for survival analysis. Benjamini-Hochberg procedure was used to adjust for multiple comparisons. Results: Among 1986 patients included, 341 (17.2%) in the 65 group had similar baseline characteristics. Outcomes: Patients in the 65 groups (15.5 vs 20.8 months, p=0.004) and shorter median PFS compared to the 50-65 (8.1 vs 9.4 month, p=0.039) and >65 groups (8.1 vs 8.6 months, p=0.07) in study 1. Findings were confirmed in study 2. Toxicity: Compared to other age groups, the
- Published
- 2022
- Full Text
- View/download PDF
41. Preclinical and Clinical Advances of Targeted Protein Degradation as a Novel Cancer Therapeutic Strategy: An Oncologist Perspective
- Author
-
He Yin, Jason B. Fleming, Richard D. Kim, Hao Xie, and Xinrui Yang
- Subjects
0301 basic medicine ,Cancer Research ,Protein degradation ,Bioinformatics ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Neoplasms ,Medicine ,Animals ,Humans ,Pharmacology (medical) ,Cellular proteins ,Therapeutic strategy ,Oncologists ,business.industry ,Cancer ,medicine.disease ,Clinical Practice ,030104 developmental biology ,Oncology ,Clinical evidence ,Reduced toxicity ,030220 oncology & carcinogenesis ,Proteolysis ,business - Abstract
PROteolysis Targeting Chimeras (PROTACs) are a family of heterobifunctional small molecules that specifically target cellular proteins for degradation. Given that their mode of action is distinct from that of small-molecule inhibitors widely used in clinical practice, PROTACs have the potential to improve current cancer therapies. Multiple studies have suggested that PROTACs exhibit enhanced pharmacodynamics and reduced toxicity both in vitro and in vivo compared to clinically relevant small-molecule kinase inhibitors. In addition, PROTACs have been reported to be less prone to mutation-mediated drug resistance in specific disease settings. Since its development in 2001, the field of targeted protein degradation, in which PROTACs are used, has expanded rapidly. However, earlier studies focused on the advancement of the technology itself, while preclinical and clinical data on the disease-modifying effect of PROTACs have only recently been reported. As preclinical and clinical evidence accumulates, the efficacy of PROTACs as targeted therapeutics-distinct from that of small-molecule kinase inhibitors-suggests potential translational benefit in the clinical setting. In this short review, we aim to describe translational potentials of PROTACs. We offer our perspectives as practicing oncologists on the preclinical and clinical data on PROTACs as novel therapeutics for both solid and hematological malignancies.
- Published
- 2020
42. Modeling precision genomic-based radiation dose response in rectal cancer
- Author
-
Michael J. Schell, Sophie Dessureault, Jessica M. Frakes, Seth Felder, Sarah E. Hoffe, Marissa Frazer, Iman Imanirad, Richard D. Kim, Javier F. Torres-Roca, Julian Sanchez, Zhigang Yuan, Kamran Ahmed, and Syeda Mahrukh Hussnain Naqvi
- Subjects
0301 basic medicine ,Adult ,Male ,Cancer Research ,medicine.medical_specialty ,Colorectal cancer ,Radiation Tolerance ,03 medical and health sciences ,0302 clinical medicine ,Radiosensitivity Index ,medicine ,Dose escalation ,Odds Ratio ,Humans ,Radiosensitivity ,Neoplasm Metastasis ,Precision Medicine ,Complete response ,Aged ,Neoplasm Staging ,Aged, 80 and over ,business.industry ,Rectal Neoplasms ,Gene Expression Profiling ,Radiation dose ,Dose-Response Relationship, Radiation ,Radiotherapy Dosage ,General Medicine ,Genomics ,Middle Aged ,medicine.disease ,Combined Modality Therapy ,030104 developmental biology ,Treatment Outcome ,Oncology ,Prospective trial ,030220 oncology & carcinogenesis ,Risk stratification ,Female ,Radiology ,Neoplasm Grading ,business ,Transcriptome - Abstract
Aim: Genomic-based risk stratification to personalize radiation dose in rectal cancer. Patients & methods: We modeled genomic-based radiation dose response using the previously validated radiosensitivity index (RSI) and the clinically actionable genomic-adjusted radiation dose. Results: RSI of rectal cancer ranged from 0.19 to 0.81 in a bimodal distribution. A pathologic complete response rate of 21% was achieved in tumors with an RSI
- Published
- 2020
43. A Phase 2 Multi-institutional Study of Nivolumab for Patients With Advanced Refractory Biliary Tract Cancer
- Author
-
Jun-Min Zhou, Bassell F. El-Rayes, Richard D. Kim, Olatunji B. Alese, Amit Mahipal, Daneng Li, Taymeyah Al-Toubah, Michael J. Schell, Baek Hui Kim, Dae-Won Kim, and Vincent Chung
- Subjects
Adult ,Male ,Cancer Research ,medicine.medical_specialty ,Population ,Phases of clinical research ,Gastroenterology ,B7-H1 Antigen ,03 medical and health sciences ,0302 clinical medicine ,Antineoplastic Agents, Immunological ,Internal medicine ,Clinical endpoint ,Biomarkers, Tumor ,Medicine ,Humans ,030212 general & internal medicine ,Adverse effect ,education ,Immune Checkpoint Inhibitors ,Original Investigation ,Aged ,Aged, 80 and over ,education.field_of_study ,business.industry ,Incidence (epidemiology) ,Hazard ratio ,Middle Aged ,Alkaline Phosphatase ,Clinical trial ,Biliary Tract Neoplasms ,Nivolumab ,Treatment Outcome ,Oncology ,Drug Resistance, Neoplasm ,030220 oncology & carcinogenesis ,Female ,business ,Hyponatremia - Abstract
IMPORTANCE: Currently, there is no established second-line systemic treatment for biliary tract cancer (BTC). Preclinical data have demonstrated that the presence of tumor-infiltrating CD8 T cells and programmed cell death 1 ligand 1–expressing tumor cells in the tumor microenvironment of BTC supports the rationale of using programmed cell death 1 protein blockade immunotherapy in BTC. OBJECTIVE: To evaluate anticancer activity of nivolumab in patients with advanced refractory BTC. DESIGN, SETTING, AND PARTICIPANTS: In this single-group, multicenter phase 2 study of nivolumab, 54 patients with histologically confirmed BTC whose disease progressed while undergoing treatment with at least 1 line but no more than 3 lines of systemic therapy were enrolled between October 5, 2016, and December 26, 2018. Analysis was performed on an intention-to-treat basis. INTERVENTIONS: Nivolumab, 240 mg, was delivered intravenously every 2 weeks for 16 weeks, and then 480 mg was delivered intravenously every 4 weeks until disease progression or unacceptable toxic effects occurred. MAIN OUTCOMES AND MEASURES: The primary end point was investigator-assessed objective response rate, and the secondary end points were progression-free survival, overall survival, and incidence of adverse events. RESULTS: A total of 54 patients (27 men and 27 women; median age, 65 years [range, 28-86 years]) enrolled, and 46 (22 men and 24 women; median age, 65 years [range, 28-86 years]) were examined for objective response with radiologic imaging. The investigator-assessed objective response rate was 22% (10 of 46), including 1 unconfirmed partial response, with a disease control rate of 59% (27 of 46). Central independent review found an objective response rate of 11% (5 of 46), including 1 unconfirmed partial response, with a disease control rate of 50% (23 of 46). All patients who responded to treated (hereafter referred to as responders) had mismatch repair protein–proficient tumors. The median duration of investigator-assessed response was not reached, with a median follow-up of 12.4 months. Among the intention-to-treat population, median progression-free survival was 3.68 months (95% CI, 2.30-5.69 months) and median overall survival was 14.24 months (95% CI, 5.98 months to not reached). Programmed cell death 1 ligand 1 expression in tumors was associated with prolonged progression-free survival (hazard ratio, 0.23; 95% CI, 0.10-0.51; P
- Published
- 2020
44. Determining Optimal Follow-up for Patients With Anal Cancer Following Chemoradiation
- Author
-
Iman Imanirad, Sophie Dessureault, George Yang, Richard D. Kim, Seth Felder, Estrella Carballido, Marissa Frazer, Jessica M. Frakes, Jordan McDonald, Sarah E. Hoffe, and Julian Sanchez
- Subjects
Adult ,Male ,Cancer Research ,medicine.medical_specialty ,Anal Carcinoma ,medicine.medical_treatment ,Aftercare ,Disease ,03 medical and health sciences ,0302 clinical medicine ,Quality of life ,Internal medicine ,medicine ,Anal cancer ,Humans ,030212 general & internal medicine ,Stage (cooking) ,Aged ,Retrospective Studies ,Aged, 80 and over ,business.industry ,Retrospective cohort study ,Chemoradiotherapy ,Middle Aged ,medicine.disease ,Anus Neoplasms ,Radiation therapy ,Oncology ,030220 oncology & carcinogenesis ,Toxicity ,Female ,Neoplasm Recurrence, Local ,business - Abstract
Background US health care is increasingly defined by over expenditure and inefficiency. Optimizing patient follow-up is critical, especially in cancers treated with high control rates. To optimize patient care, this study assessed time to disease recurrence or toxicity in patients with anal carcinoma. Materials and methods In total, 140 patients with biopsy-proven, nonmetastatic anal carcinoma, treated with chemoradiation utilizing intensity-modulated radiation therapy, were identified from our institutional database. This retrospective study evaluated local recurrence (LR), distant metastasis (DM), overall survival (OS), and late ≥grade 3 toxicity (LG3T). Patients were followed posttreatment every 3 months for 2 years, every 6 months in years 3 to 5, then yearly thereafter per NCCN recommendations. Results The median age and follow-up was 58 years and 27 months, respectively. Patients were categorized into high (n=61; 44%) and low (n=77; 55%) risk groups based on stage. The 2-year LC, DMFS, and OS were 93%, 94%, and 89% and 5-year LC, DMFS, OS were 92%, 87%, and 85%, respectively. Overall, there were 29 events (9 LR, 11 DM, and 9 LG3T), with 62% of events occurring within year 1 and 79% within 2 years. Stratified by event type, at 2 years 89% of LR, 64% of DM, and 89% LG3T were identified. At the remaining follow-up points, the event incidence rate was 1.3%. Conclusion With the majority of recurrences/toxicities occurring within the first 2 years, a reduction in follow-up during years 3 to 5 may provide adequate surveillance. Revisions of the current recommendations could maximize resources while improving patient quality of life.
- Published
- 2020
45. CT-based radiomic features to predict pathological response in rectal cancer: A retrospective cohort study
- Author
-
Richard D. Kim, Seth Felder, Zhigang Yuan, Marissa Frazer, Kujtim Latifi, Julian Sanchez, Iman Imanirad, Geoffrey Zhang, Jessica M. Frakes, Eduardo G. Moros, Sarah E. Hoffe, Louis B. Harrison, Sophie Dessureault, and Vladimir Feygelman
- Subjects
Oncology ,Adult ,Male ,medicine.medical_specialty ,Tumour regression ,Imaging biomarker ,Colorectal cancer ,Pathological response ,030218 nuclear medicine & medical imaging ,Machine Learning ,03 medical and health sciences ,0302 clinical medicine ,Predictive Value of Tests ,Internal medicine ,medicine ,Biomarkers, Tumor ,Humans ,Radiology, Nuclear Medicine and imaging ,Pathological ,Complete response ,Aged ,Neoplasm Staging ,Retrospective Studies ,Aged, 80 and over ,business.industry ,Rectal Neoplasms ,Retrospective cohort study ,Exploratory analysis ,Chemoradiotherapy ,Middle Aged ,medicine.disease ,Neoadjuvant Therapy ,030220 oncology & carcinogenesis ,Florida ,Female ,Neoplasm Grading ,business ,Tomography, X-Ray Computed - Abstract
Introduction Innovative biomarkers to predict treatment response in rectal cancer would be helpful in optimizing personalized treatment approaches. In this study, we aimed to develop and validate a CT-based radiomic imaging biomarker to predict pathological response. Methods We used two independent cohorts of rectal cancer patients to develop and validate a CT-based radiomic imaging biomarker predictive of treatment response. A total of 91 rectal cancer cases treated from 2009 to 2018 were assessed for the tumour regression grade (TRG) (0 = pathological complete response, pCR; 1 = moderate response; 2 = partial response; 3 = poor response). Exploratory analysis was performed by combining pre-treatment non-contrast CT images and patterns of TRG. The models built from the training cohort were further assessed using the independent validation cohort. Results The patterns of pathological response in training and validation groups were TRG 0 (n = 14, 23.3%; n = 6, 19.4%), 1 (n = 31, 51.7%; n = 15, 48.4%), 2 (n = 12, 20.0%; n = 7, 22.6%) and 3 (n = 3, 5.0%; n = 3, 9.7%), respectively. Separate predictive models were built and analysed from CT features for pathological response. For pathological response prediction, the model including 8 radiomic features by random forest method resulted in 83.9% accuracy in predicting TRG 0 vs TRG 1-3 in validation. Conclusion The pre-treatment CT-based radiomic signatures were developed and validated in two independent cohorts. This imaging biomarker provided a promising way to predict pCR and select patients for non-operative management.
- Published
- 2020
46. Multidisciplinary Management of Liver, Pancreatic, and Gastric Malignancies in Older Adults
- Author
-
Nadia Saeed, Richard D. Kim, Kaldhoun Almhanna, Amit Mahipal, Danielle K. DePeralta, J.M. Frakes, and Daniel A. Anaya
- Subjects
Oncology ,medicine.medical_specialty ,business.industry ,Multidisciplinary approach ,Hepatocellular carcinoma ,Internal medicine ,medicine ,medicine.disease ,business - Published
- 2020
- Full Text
- View/download PDF
47. Core homologous recombination mutations and improved survival in nonpancreatic GI cancers
- Author
-
Elaine Tan, Junmin Whiting, Christine Marie Walko, Todd C Knepper, Hao Xie, Iman Imanirad, Estrella M. Carballido, Seth Felder, Jessica M. Frakes, Qianxing Mo, Jennifer Permuth, Richard D. Kim, Daniel A. Anaya, Jason B. Fleming, and Ibrahim Halil Halil Sahin
- Subjects
Cancer Research ,Oncology - Abstract
663 Background: GI malignancies continue to be a major cause of cancer related deaths, despite advances in therapy options. In pancreatic cancer, the presence of homologous recombination mutations (HRM) has been shown to confer increased sensitivity to platinum chemotherapy. However, the role of HRM mutations in other GI cancers remains to be determined. The focus of this study is to evaluate the prognostic nature of core and noncore HRM in nonpancreatic GI cancers. Additionally, we aim to understand the predictive nature of these mutations related to platinum exposure. Methods: This was a retrospective review of patients at Moffitt Cancer Center with a primary stage IV nonpancreatic GI cancer treated with platinum therapy. All patients had next generation sequencing and were in the Clinical Genomics Action Committee database between 1/1/2013 and 11/1/2020. All patients had either a core HRM (BRCA1, BRCA2, PALB2) or noncore HRM (such as ATM, ATR, BARD1, BRIP1, FANCA, NBN, and RAD51). Patients were grouped into core HRM vs. noncore HRM. Response was stratified between responders (complete response and partial response) vs. non responders (stable disease and progressive disease), based on RECIST criteria. Progression free survival and overall survival were estimated using Kaplan-Meier method, and the log-rank test was used to assess the difference in progression free survival (PFS) and overall survival (OS) by HRM status and type of platinum therapy used. Results: There were 72 patients included in the study: the majority of patients were male (65.3%) and Caucasian (87.5%). Twenty-one (29.2%) patients had a core HRM and 51 (70.8%) had a noncore HRM. There was no significant difference in response rate between patients with core HRM and patients with noncore HRM for evaluable patients (n = 66): 20.0% of patients with core HRM vs. 21.7% of patients with noncore HRM demonstrated a response to platinum therapy (p = 0.41). An improved OS was noted for patients with core HRM vs. those with noncore HRM at 68.9 vs. 24.3 months (p = 0.019). An improved PFS was also seen in patients with core HRM at 10.4 months vs. 6.3 months with noncore HRM (p = 0.027). There was no difference in PFS based on type of platinum therapy used (oxaliplatin vs. carboplatin vs. cisplatin), while an improved OS was noted for patients treated with oxaliplatin vs. carboplatin vs. cisplatin at 43.1 vs. 28.2 vs. 16.0 months (p = 0.011). Conclusions: Our study demonstrated that in stage IV nonpancreatic GI malignancies treated with platinum therapy, patients with core HRM had a greater OS and PFS compared to those with noncore HRM, suggesting a potential prognostic and predictive role of these mutations. Further studies are needed to confirm our findings.
- Published
- 2022
- Full Text
- View/download PDF
48. A phase II study of TAS-102 (FTD/TPI) in combination with ramucirumab (RAM) in advanced, refractory gastric (GC) or gastroesophageal junction (GEJ) adenocarcinomas (GEAs)
- Author
-
Rutika Mehta, Richard D. Kim, Maria E Martinez Jimenez, Kirsten Blue, Trenton Avriett, Emily Kelbert, Kara Miller, Christopher Ray, Tiffany Valone, Woojoo Lee, Youngchul Kim, and Dae Won Kim
- Subjects
Cancer Research ,Oncology - Abstract
302 Background: The RAINBOW trial established the standard of care for treatment of metastatic GEAs with ramucirumab and paclitaxel after failure of fluoropyrimidine and platinum-based chemotherapy. While the combination achieved an objective response rate (ORR) of 28%, the incidence of any grade neuropathy was 46%. Therefore, there is an unmet need for novel treatment combinations that minimize the long-term toxicity of neuropathy. In a recently published Asian study, the combination of RAM+TAS-102 showed good disease control and acceptable toxicity profile. Methods: This was a single arm, single institution phase II trial using the combination of TAS-102 plus RAM in refractory GEAs. Patients (pts) received RAM 8mg/kg intravenously on day 1 and 15, and TAS-102 35mg/m2 orally twice daily on days 1-5 and days 8-12 every 28-day cycle. The primary endpoint was 6-months overall survival (OS) rate and secondary endpoints were progression free survival (PFS), ORR and safety profile. The trial was registered at www.clinicaltrials.gov (NCT03686488). Results: At data cut-off of August 15, 2021, 23 pts were enrolled. Baseline demographics are as follows: median age of 62 years (range: 23-74), median lines of prior therapy of 1 (1: 14 pts vs ≥2: 9 pts) and location of primary tumor (GEJ:19 vs GC: 4). 6-month OS rate was 56.2%. Median OS was 6.2 month (95% CI: 5.4-7.0) and median PFS was 4.9 months with median observation of 2.3 months. Of 17 evaluable pts defined as more than one baseline imaging, 1 (6%) had partial response (PR) and 15 (88%) had stable disease (RECIST v1.1). Eleven pts came off the study due to progression of disease, 8 for toxicities and 4 for consent withdrawal. Most common treatment-emergent adverse events (TEAEs) were diarrhea (39%), fatigue (39%) and hypertension (39%). Total 11 pts (48%) experienced grade 3 and 4 TEAEs, and most common Gr3 and 4 TEAEs were neutropenia (17%) and anemia (13%). Conclusions: The combination of RAM and TAS-102 showed very similar disease control rate as the study in Asia. The combination has now shown modest activity in advanced GEAs and should be investigated further now in the context of patients receiving immunotherapy-based treatment as first-line treatment. A randomized phase II study is currently enrolling patients with ramucirumab and TAS-102 or paclitaxel. Clinical trial information: NCT03686488.
- Published
- 2022
- Full Text
- View/download PDF
49. Association of BRAF V600E mutation with survival in patients with metastatic mismatch repair-deficient colorectal cancer
- Author
-
Elaine Tan, Junmin Whiting, Hao Xie, Iman Imanirad, Estrella M. Carballido, Seth Felder, Jessica M. Frakes, Qianxing Mo, Christine Marie Walko, Jennifer Permuth, Richard D. Kim, Daniel A. Anaya, Jason B. Fleming, and Ibrahim Halil Halil Sahin
- Subjects
Cancer Research ,Oncology ,neoplasms ,digestive system diseases - Abstract
174 Background: Colorectal cancer (CRC), while one of the most common cancer diagnoses, can behave heterogeneously based on molecular characteristics. A subset of patients (pts) with CRC are characterized with mismatch repair deficiency (MMR-d), these pts exhibit encouraging responses to immunotherapy. The predictive nature of various factors, such as BRAF status, age, and MMR-D protein loss type, have been investigated in pts with MMR-d CRC. However, the prognostic role of these factors has not been well established. The purpose of this study was to identify characteristics that influence survival in MMR-d mCRC. Methods: This study evaluated pts with MMR-d mCRC in the Flatiron database. Overall survival (OS) was determined from date of diagnosis of stage IV disease to date of death and stratified based on age greater than or less than 50 years, BRAF mutation status, RAS mutation status, and type of MMR gene loss. For statistical analysis, the Chi-Square test was implemented to determine the prognostic significance of clinical and molecular features. Univariate and multivariate analyses were determined through the Cox regression model. Results: There were 1,101 pts in the study. The majority of pts were older than 50 (79.7%), Caucasian (75%), and had ECOG 0-1 (83.4%). Among the 803 pts with known BRAF status, 44.3% (n=356) had BRAF V600E mutation and 55.7% (n=447) were BRAF wildtype. Pts with BRAF V600E mutation had OS of 18.9 months vs. 33.2 months for pts with wild type BRAF (HR 1.52, 95% CI: 1.25-1.86, p
- Published
- 2022
- Full Text
- View/download PDF
50. Multicenter, randomized, double‐blind phase 2 trial of <scp>FOLFIRI</scp> with regorafenib or placebo as second‐line therapy for metastatic colorectal cancer
- Author
-
Ray McDermott, Samer S. Kasbari, George Hosni Yacoub, William C. Zamboni, William Grogan, Anastasia Ivanova, Bassel F. El-Rayes, Theresa Ryan, Olugbenga Olowokure, Allen Lee Cohn, Dominic T. Moore, Bert H. O'Neil, Tanios Bekaii-Saab, Seamus O'Reilly, Nishan H. Fernando, John McCaffrey, Richard D. Kim, Hanna K. Sanoff, Richard M. Goldberg, Anne M. Horgan, Gregory D. Leonard, and Gary Bradley Sherrill
- Subjects
Adult ,Male ,0301 basic medicine ,Cancer Research ,medicine.medical_specialty ,Drug-Related Side Effects and Adverse Reactions ,Bevacizumab ,Pyridines ,Leucovorin ,Kaplan-Meier Estimate ,Irinotecan ,Gastroenterology ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Double-Blind Method ,Multicenter trial ,Internal medicine ,Regorafenib ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Humans ,Aged ,Aflibercept ,business.industry ,Phenylurea Compounds ,Middle Aged ,medicine.disease ,Progression-Free Survival ,Oxaliplatin ,030104 developmental biology ,Oncology ,chemistry ,030220 oncology & carcinogenesis ,FOLFIRI ,Camptothecin ,Female ,Fluorouracil ,Colorectal Neoplasms ,business ,Febrile neutropenia ,medicine.drug - Abstract
Background Regorafenib, a multikinase inhibitor that inhibits angiogenesis, growth, and proliferation, prolongs survival as monotherapy in patients with refractory colorectal cancer. This international, double-blind, placebo-controlled, multicenter trial assessed the efficacy of regorafenib with folinic acid, fluorouracil, and irinotecan (FOLFIRI) as a second-line treatment for metastatic colorectal cancer. Methods Patients with metastatic colorectal cancer who progressed on first-line oxaliplatin and fluoropyrimidine enrolled at 45 sites in the United States and Ireland. Patients, stratified by prior bevacizumab use, were randomized 2:1 to regorafenib or placebo. The treatment consisted of FOLFIRI on days 1 and 2 and days 15 and 16 with 160 mg of regorafenib or placebo on days 4 to 10 and days 18 to 24 of every 28-day cycle. Crossover was not allowed. The primary endpoint was progression-free survival (PFS). Under the assumption of a 75% event rate, 180 patients were required for 135 events to achieve 90% power to detect a hazard ratio (HR) of 0.65 with a 1-sided α value of .1. Results One hundred eighty-one patients were randomized (120 to regorafenib-FOLFIRI and 61 to placebo-FOLFIRI) with a median age of 62 years. Among these, 117 (65%) received prior bevacizumab or aflibercept. PFS was longer with regorafenib-FOLFIRI than placebo-FOLFIRI (median, 6.1 vs 5.3 months; HR, 0.73; 95% confidence interval [CI], 0.53-1.01; log-rank P = .056). The median overall survival was not longer (HR, 1.01; 95% CI, 0.71-1.44). The response rate was higher with regorafenib-FOLFIRI (34%; 95% CI, 25%-44%) than placebo-FOLFIRI (21%; 95% CI, 11%-33%; P = .07). Grade 3/4 adverse events with a >5% absolute increase from regorafenib included diarrhea, neutropenia, febrile neutropenia, hypophosphatemia, and hypertension. Conclusions The addition of regorafenib to FOLFIRI as second-line therapy for metastatic colorectal cancer only modestly prolonged PFS over FOLFIRI alone. Cancer 2018. © 2018 American Cancer Society.
- Published
- 2018
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.