201
|
Pathak A, Pal AK, Roy S, Nandave M, Jain K. Role of Angiogenesis and Its Biomarkers in Development of Targeted Tumor Therapies. Stem Cells Int 2024; 2024:9077926. [PMID: 38213742 PMCID: PMC10783989 DOI: 10.1155/2024/9077926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 11/21/2023] [Accepted: 12/04/2023] [Indexed: 01/13/2024] Open
Abstract
Angiogenesis plays a significant role in the human body, from wound healing to tumor progression. "Angiogenic switch" indicates a time-restricted event where the imbalance between pro- and antiangiogenic factors results in the transition from prevascular hyperplasia to outgrowing vascularized tumor, which eventually leads to the malignant cancer progression. In the last decade, molecular players, i.e., angiogenic biomarkers and underlying molecular pathways involved in tumorigenesis, have been intensely investigated. Disrupting the initiation and halting the progression of angiogenesis by targeting these biomarkers and molecular pathways has been considered as a potential treatment approach for tumor angiogenesis. This review discusses the currently known biomarkers and available antiangiogenic therapies in cancer, i.e., monoclonal antibodies, aptamers, small molecular inhibitors, miRNAs, siRNAs, angiostatin, endostatin, and melatonin analogues, either approved by the U.S. Food and Drug Administration or currently under clinical and preclinical investigations.
Collapse
Affiliation(s)
- Anchal Pathak
- Drug Delivery and Nanomedicine Research Laboratory, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER)-Raebareli, Lucknow, India
| | - Ajay Kumar Pal
- Department of Pharmacology, Delhi Pharmaceutical Sciences and Research University (DPSRU), New Delhi 110017, India
| | - Subhadeep Roy
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Kolkata, West Bengal, India
| | - Mukesh Nandave
- Department of Pharmacology, Delhi Pharmaceutical Sciences and Research University (DPSRU), New Delhi 110017, India
| | - Keerti Jain
- Drug Delivery and Nanomedicine Research Laboratory, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER)-Raebareli, Lucknow, India
| |
Collapse
|
202
|
Qu W, Liu Z, Chen X, Liu B, Zhao Y, Yan H, Qu X, Li S, Zang A, Sun Y, Zhu L, Zhou A. Regorafenib monotherapy or combined with an immune-checkpoint inhibitor as later-line treatment for metastatic colorectal cancer: a multicenter, real-world retrospective study in China. BMC Cancer 2024; 24:22. [PMID: 38166647 PMCID: PMC10763390 DOI: 10.1186/s12885-023-11700-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2023] [Accepted: 11/30/2023] [Indexed: 01/05/2024] Open
Abstract
OBJECTIVE To evaluate the efficacy and safety of regorafenib monotherapy or in combination with immune-checkpoint inhibitor while treating Chinese patients with metastatic colorectal cancer (mCRC): a real-world study. METHODS The data of patients with metastatic colorectal cancer who received regorafenib-containing regimen as the third or later line treatment at ten Chinese hospitals from Aug 2017 to Jun 2020 were retrospectively analyzed, including dosing details, survival data as well as adverse events. Survival analysis was further performed for patients administrated with regorafenib monotherapy and combined with an immune-checkpoint inhibitor based on Kaplan-Meier and Cox regression methods. The primary endpoint was overall survival. RESULTS A total of 537 patients were included with a median age of 61, among whom 376 received regorafenib monotherapy and 245 received regorafenib combined with immune-checkpoint inhibitors. The clinicopathological characteristics of the two groups at baseline were mainly balanced. No significant difference in progression-free survival (PFS) was observed in patients receiving regorafenib monotherapy or combination therapy (3.8 vs. 5.5 months, p = 0.170). In contrast, patients receiving combination therapy had a more prolonged overall survival (OS) than those receiving regorafenib monotherapy (13.5 vs. 10.0 months, p = 0.001). The treatment regimen and regorafenib dosage were significant prognostic factors in the multivariate analysis. Significant benefits in PFS and OS were achieved in KRAS mutant and anti-angiogenesis treatment-naïve subgroups receiving combination therapy compared to monotherapy. No apparent increase was recorded in treatment-related adverse events in patients receiving combination therapy. CONCLUSION Regorafenib plus an immune-checkpoint inhibitor has already been a widely adopted strategy in the later-line treatment for mCRC in the real world. The combination therapy yielded a significantly prolonged overall survival than regorafenib alone, with a manageable safety profile in Chinese patients, and warrants further investigation. TRIAL REGISTRATION ClinicalTrials.gov Identifier: NCT04835324. Registered 6th April 2021.
Collapse
Affiliation(s)
- Wang Qu
- Department of Medical Oncology, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Zimin Liu
- Department of Medical Oncology, Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Xiaobing Chen
- Department of Medical Oncology, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, 450003, China
| | - Bo Liu
- Department of Medical Oncology, Shandong Cancer Hospital, Jinan, 250117, China
| | - YunBo Zhao
- Department of Oncology, Beijing Hospital, National Center of Gerontology, Beijing, 100730, China
| | - Hao Yan
- Department of Oncology, Tianjin Union Medical Center, Tianjin, 300122, China
| | - Xiujuan Qu
- Department of Oncology, The First Hospital of China Medical University, Shenyang, 110001, China
| | - Shengmian Li
- Department of Gastroenterology and Hepatology, Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, China
| | - Aimin Zang
- Department of Oncology, Affiliated Hospital of Hebei University, Baoding, 071000, China
| | - Yongkun Sun
- Department of Medical Oncology, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Liangjun Zhu
- Department of Medical Oncology, Jiangsu Cancer Hospital, NO,42 Bai Zi Ting, Xuanwu District, Nanjing, 210000, China.
| | - Aiping Zhou
- Department of Medical Oncology, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
203
|
Taşçı EŞ, Oyan B, Sönmez Ö, Mutlu AU, Atcı MM, Sakin A, Öner İ, Çınkır HY, Eryılmaz MK, Çağlayan D, Balçık OY, Paksoy N, Karabulut S, Salim DK, Bilir C, Özen M, Özçelik M, Arıcan A, Akagündüz B, İnal A, Aydın D, Özer L, Gülmez A, Turhal NS, Esen SA, Algın E, Akbaş S, İriağaç Y, Şakalar T, Ünal Ç, Er Ö, Seçmeler Ş, Bozkurt M. Comparing the efficacy of regorafenib and 5-fluorouracil-based rechallenge chemotherapy in the third-line treatment of metastatic colorectal cancer. BMC Cancer 2024; 24:16. [PMID: 38166764 PMCID: PMC10763265 DOI: 10.1186/s12885-023-11783-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 12/21/2023] [Indexed: 01/05/2024] Open
Abstract
BACKGROUND The optimal treatment for metastatic colorectal cancer (mCRC) after the second line is still controversial. Regorafenib has been the standard of care in this setting as it improved overall survival (OS) compared to placebo. In real-world practice chemotherapy rechallenge is also a preferred option even though supporting evidence is not enough. We aim to compare the efficacy of regorafenib and 5-fluorouracil-based (5-FU) rechallenge treatment in the third line setting of mCRC. METHODS In this retrospective multi-institutional trial, mCRC patients from 21 oncology centers who progressed after 2 lines of chemotherapy were analyzed. Patients who were treated with regorafenib or rechallenge therapy in the third-line setting were eligible. Rechallenge chemotherapy was identified as the re-use of the 5-FU based regimen which was administered in one of the previous treatment lines. OS, disease control rate (DCR), progression free survival (PFS) and toxicity were analyzed. RESULTS Three hundred ninety-four mCRC patients were included in the study. 128 (32.5%) were in the rechallenge, and 266 (67.5%) were in the regorafenib group. Median PFS was 5.82 months in rechallenge and 4 months in regorafenib arms (hazard ratio:1.45,95% CI, p = 0.167). DCR was higher in the rechallenge group than regorafenib (77% vs 49.5%, respectively, p = < 0.001). Median OS after the third-line treatment was 11.99 (95% CI, 9.49-14.49) and 8.08 months (95% CI, 6.88-9.29) for rechallenge and regorafenib groups, respectively (hazard ratio:1.51, 95% CI, p < 0.001). More adverse effects and discontinuation were seen with regorafenib treatment. CONCLUSION Our study revealed that higher disease control and OS rates were achieved with rechallenge treatment compared to regorafenib, especially in patients who achieved disease control in one of the first two lines of therapy.
Collapse
Affiliation(s)
- Elif Şenocak Taşçı
- Department of Medical Oncology, Saglık Bilimleri University, Kanuni Sultan Süleyman Research and Training Hospital, Istanbul, Turkey.
| | - Başak Oyan
- Department of Medicine, Acıbadem MAA University, Istanbul, Turkey
| | - Özlem Sönmez
- Department of Medicine, Acıbadem MAA University, Istanbul, Turkey
| | - Arda Ulaş Mutlu
- Department of Medicine, Acıbadem MAA University, Istanbul, Turkey
| | - Muhammed Mustafa Atcı
- Department of Medical Oncology, Haseki Education and Research Hospital, Istanbul, Turkey
| | - Abdullah Sakin
- Department of Medical Oncology, Medipol Bahçelievler Hospital, Istanbul, Turkey
| | - İrem Öner
- Department of Medical Oncology, Konya City Hospital, Konya, Turkey
| | - Havva Yeşil Çınkır
- Department of Medical Oncology, Faculty of Medicine, Gaziantep University, Gaziantep, Turkey
| | - Melek Karakurt Eryılmaz
- Meram Faculty of Medicine, Department of Medical Oncology, Necmettin Erbakan University, Konya, Turkey
| | - Dilek Çağlayan
- Meram Faculty of Medicine, Department of Medical Oncology, Necmettin Erbakan University, Konya, Turkey
| | - Onur Yazdan Balçık
- Department of Medical Oncology, Mardin Education and Research Hospital, Mardin, Turkey
| | - Nail Paksoy
- Department of Medical Oncology, Tekirdağ Dr. İsmail Fehmi Cumalıoğlu City Hospital, Tekirdağ, Turkey
| | - Senem Karabulut
- Department of Medical Oncology, Şişli Kolan Hospital, Istanbul, Turkey
| | - Derya Kıvrak Salim
- Department of Medical Oncology, Antalya Education and Research Hospital, Antalya, Turkey
| | - Cemil Bilir
- Department of Medical Oncology, Medical Park Hospital, Istanbul, Turkey
| | - Miraç Özen
- Department of Medical Oncology, Sakarya University Research and Education Hospital, Sakarya, Turkey
| | - Melike Özçelik
- Department of Medical Oncology, University of Health Sciences Umraniye Education and Research Hospital, Istanbul, Turkey
| | - Ali Arıcan
- Department of Medicine, Acıbadem MAA University, Istanbul, Turkey
| | - Baran Akagündüz
- Department of Medical Oncology, Binali Yıldırım University, Erzincan, Turkey
| | - Ali İnal
- Department of Medical Oncology, Mersin City Hospital, Mersin, Turkey
| | - Dinçer Aydın
- Department of Medical Oncology, Derince Education and Research Hospital, Kocaeli, Turkey
| | - Leyla Özer
- Department of Medicine, Acıbadem MAA University, Istanbul, Turkey
| | - Ahmet Gülmez
- Department of Medical Oncology, Adana City Hospital, Adana, Turkey
| | | | - Selin Aktürk Esen
- Department of Medical Oncology, Ankara City Hospital, Ankara, Turkey
| | - Efnan Algın
- Department of Medical Oncology, Ankara City Hospital, Ankara, Turkey
| | - Sinem Akbaş
- Department of Medical Oncology, Koç University Hospital, Istanbul, Turkey
| | - Yakup İriağaç
- Department of Medical Oncology, Namık Kemal University, Tekirdağ, Turkey
| | - Teoman Şakalar
- Department of Medical Oncology, Necip Fazıl City Hospital, Kahramanmaraş, Turkey
| | - Çağlar Ünal
- Department of Medical Oncology, Bilim University, Istanbul, Turkey
| | - Özlem Er
- Department of Medicine, Acıbadem MAA University, Istanbul, Turkey
| | - Şaban Seçmeler
- Department of Medical Oncology, Medical Park Bahçelievler Hospital, Istanbul, Turkey
| | - Mustafa Bozkurt
- Department of Medicine, Acıbadem MAA University, Istanbul, Turkey
| |
Collapse
|
204
|
Soler-González G, Sastre-Valera J, Viana-Alonso A, Aparicio-Urtasun J, García-Escobar I, Gómez-España MA, Guillén-Ponce C, Molina-Garrido MJ, Gironés-Sarrió R. Update on the management of elderly patients with colorectal cancer. Clin Transl Oncol 2024; 26:69-84. [PMID: 37498507 PMCID: PMC10761480 DOI: 10.1007/s12094-023-03243-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 05/31/2023] [Indexed: 07/28/2023]
Abstract
Colorectal cancer (CRC) is one of the most common tumours worldwide, and 70% of CRC patients are over 65 years of age. However, the scientific evidence available for these patients is poor, as they are underrepresented in clinical trials. Therefore, a group of experts from the Oncogeriatrics Section of the Spanish Society of Medical Oncology (SEOM), the Spanish Cooperative Group for the Treatment of Digestive Tumours, (TTD) and the Multidisciplinary Spanish Group of Digestive Cancer (GEMCAD) have reviewed the scientific evidence available in older patients with CRC. This group of experts recommends a multidisciplinary approach and geriatric assessment (GA) before making a therapeutic decision because GA predicts the risk of toxicity and survival and helps to individualize treatment. In addition, elderly patients with localized CRC should undergo standard cancer resection, preferably laparoscopically. The indication for adjuvant chemotherapy (CT) should be considered based on the potential benefit, the risk of recurrence, the life expectancy and patient comorbidities. When the disease is metastatic, the possibility of radical treatment with surgery, radiofrequency (RF) or stereotactic body radiation therapy (SBRT) should be considered. The efficacy of palliative CT is similar to that seen in younger patients, but elderly patients are at increased risk of toxicity. Clinical trials should be conducted with the elderly population and include GAs and specific treatment plans.
Collapse
Affiliation(s)
- Gemma Soler-González
- Departamento de Oncología Médica, Spanish Society of Medical Oncology (SEOM) Oncogeriatrics Section, Institut Català d'Oncologia (ICO) L'Hospitalet, Avinguda de la Granvia de l'Hospitalet 199-203, L'Hospitalet de Llobregat, 08908, Barcelona, Spain.
| | - Javier Sastre-Valera
- Spanish Cooperative Group for the Treatment of Digestive Tumours (TTD), Clinico San Carlos University Hospital, Madrid, Spain
| | - Antonio Viana-Alonso
- Spanish Society of Medical Oncology (SEOM) Oncogeriatrics Section, Nuestra Señora del Prado General University Hospital, Talavera de la Reina, Spain
| | - Jorge Aparicio-Urtasun
- Multidisciplinary Spanish Group of Digestive Cancer (GEMCAD), Polytechnic la Fe University Hospital, Valencia, Spain
| | - Ignacio García-Escobar
- Spanish Society of Medical Oncology (SEOM) Oncogeriatrics Section, General University Hospital of Toledo, Toledo, Spain
| | - María Auxiliadora Gómez-España
- Spanish Society of Medical Oncology (SEOM) Oncogeriatrics Section, Reina Sofía University Hospital. Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Córdoba, Spain
| | - Carmen Guillén-Ponce
- Spanish Society of Medical Oncology (SEOM) Oncogeriatrics Section, Ramón y Cajal University Hospital, Madrid, Spain
| | - María José Molina-Garrido
- Spanish Society of Medical Oncology (SEOM) Oncogeriatrics Section, Virgen de la Luz Hospital, Cuenca, Spain
| | - Regina Gironés-Sarrió
- Spanish Society of Medical Oncology (SEOM) Oncogeriatrics Section, Polytechnic la Fe University Hospital, Valencia, Spain
| |
Collapse
|
205
|
Trembath HE, Yeh JJ, Lopez NE. Gastrointestinal Malignancy: Genetic Implications to Clinical Applications. Cancer Treat Res 2024; 192:305-418. [PMID: 39212927 DOI: 10.1007/978-3-031-61238-1_15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Advances in molecular genetics have revolutionized our understanding of the pathogenesis, progression, and therapeutic options for treating gastrointestinal (GI) cancers. This chapter provides a comprehensive overview of the molecular landscape of GI cancers, focusing on key genetic alterations implicated in tumorigenesis across various anatomical sites including GIST, colon and rectum, and pancreas. Emphasis is placed on critical oncogenic pathways, such as mutations in tumor suppressor genes, oncogenes, chromosomal instability, microsatellite instability, and epigenetic modifications. The role of molecular biomarkers in predicting prognosis, guiding treatment decisions, and monitoring therapeutic response is discussed, highlighting the integration of genomic profiling into clinical practice. Finally, we address the evolving landscape of precision oncology in GI cancers, considering targeted therapies and immunotherapies.
Collapse
Affiliation(s)
- Hannah E Trembath
- Division of Colon and Rectal Surgery, Department of Surgery, University of California San Diego, 4303 La Jolla Village Drive Suite 2110, San Diego, CA, 92122, USA
- Division of Surgical Oncology, Department of Surgery, University of North Carolina, 170 Manning Drive, CB#7213, 1150 Physician's Office Building, Chapel Hill, NC, 27599-7213, USA
| | - Jen Jen Yeh
- Division of Colon and Rectal Surgery, Department of Surgery, University of California San Diego, 4303 La Jolla Village Drive Suite 2110, San Diego, CA, 92122, USA
- Division of Surgical Oncology, Department of Surgery, University of North Carolina, 170 Manning Drive, CB#7213, 1150 Physician's Office Building, Chapel Hill, NC, 27599-7213, USA
| | - Nicole E Lopez
- Division of Colon and Rectal Surgery, Department of Surgery, University of California San Diego, 4303 La Jolla Village Drive Suite 2110, San Diego, CA, 92122, USA.
- Division of Surgical Oncology, Department of Surgery, University of North Carolina, 170 Manning Drive, CB#7213, 1150 Physician's Office Building, Chapel Hill, NC, 27599-7213, USA.
| |
Collapse
|
206
|
Kuboki Y, Fakih M, Strickler J, Yaeger R, Masuishi T, Kim EJ, Bestvina CM, Kopetz S, Falchook GS, Langer C, Krauss J, Puri S, Cardona P, Chan E, Varrieur T, Mukundan L, Anderson A, Tran Q, Hong DS. Sotorasib with panitumumab in chemotherapy-refractory KRAS G12C-mutated colorectal cancer: a phase 1b trial. Nat Med 2024; 30:265-270. [PMID: 38177853 PMCID: PMC11135132 DOI: 10.1038/s41591-023-02717-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 11/14/2023] [Indexed: 01/06/2024]
Abstract
The current third-line (and beyond) treatment options for RAS-mutant metastatic colorectal cancer have yielded limited efficacy. At the time of study start, the combination of sotorasib, a KRAS (Kirsten rat sarcoma viral oncogene homolog)-G12C inhibitor, and panitumumab, an epidermal growth factor receptor (EGFR) inhibitor, was hypothesized to overcome treatment-induced resistance. This phase 1b substudy of the CodeBreaK 101 master protocol evaluated sotorasib plus panitumumab in patients with chemotherapy-refractory KRASG12C-mutated metastatic colorectal cancer. Here, we report the results in a dose-exploration cohort and a dose-expansion cohort. Patients received sotorasib (960 mg, once daily) plus panitumumab (6 mg kg-1, once every 2 weeks). The primary endpoints were safety and tolerability. Secondary endpoints included efficacy and pharmacokinetics. Exploratory biomarkers at baseline were assessed. Forty-eight patients (dose-exploration cohort, n = 8; dose-expansion cohort, n = 40) were treated. Treatment-related adverse events of any grade and grade ≥3 occurred in 45 (94%) and 13 (27%) patients, respectively. In the dose-expansion cohort, the confirmed objective response rate was 30.0% (95% confidence interval (CI) 16.6%, 46.5%). Median progression-free survival was 5.7 months (95% CI 4.2, 7.7 months). Median overall survival was 15.2 months (95% CI 12.5 months, not estimable). Prevalent genomic coalterations included APC (84%), TP53 (74%), SMAD4 (33%), PIK3CA (28%) and EGFR (26%). Sotorasib-panitumumab demonstrated acceptable safety with promising efficacy in chemotherapy-refractory KRASG12C-mutated metastatic colorectal cancer. ClinicalTrials.gov identifier: NCT04185883 .
Collapse
Affiliation(s)
| | - Marwan Fakih
- City of Hope Comprehensive Cancer Center, Duarte, CA, USA
| | | | - Rona Yaeger
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | - Edward J Kim
- UC Davis Comprehensive Cancer Center, Sacramento, CA, USA
| | | | - Scott Kopetz
- The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | - Corey Langer
- University of Pennsylvania, Philadelphia, PA, USA
| | | | - Sonam Puri
- Huntsman Cancer Institute, Salt Lake City, UT, USA
| | | | | | | | | | | | - Qui Tran
- Amgen Inc., Thousand Oaks, CA, USA
| | - David S Hong
- The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
207
|
Tsukanov V, Ryabokon A, Tonkikh Y, Vasyutin A. Colorectal cancer: incidence, risk factors, and screening. RUSSIAN JOURNAL OF PREVENTIVE MEDICINE AND PUBLIC HEALTH 2024; 27:93. [DOI: 10.17116/profmed20242703193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/12/2024]
|
208
|
Khan IA, Singh N, Gunjan D, Dash NR, Nayak B, Gupta S, Saraya A. Elevated levels of peripheral Th17 cells and Th17-related cytokines in patients with periampullary adenocarcinoma. Hum Immunol 2024; 85:110748. [PMID: 38177009 DOI: 10.1016/j.humimm.2023.110748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 12/12/2023] [Accepted: 12/29/2023] [Indexed: 01/06/2024]
Abstract
AIM Periampullary adenocarcinoma (PAC) is a malignant tumor originating at the ampulla of Vater, distal common bile duct, head of the pancreas, ampulla and duodenum. The levels of circulating Th17 cells and Th17-related cytokines in patients with PAC remain unreported. Therefore, the aim of this study was to determine the levels of circulating Th17 cells and Th17-related cytokines in patients with PAC. MATERIALS AND METHODS Flow cytometry was used to measure Th17 cell proportions in PBMCs from 60 PAC patients and 30 healthy controls. Enzyme-linked immunosorbent assay (ELISA) was used to quantify IL-17A and IL-23 levels in serum samples, while quantitative reverse transcription polymerase chain reaction (qRT-PCR) assessed IL-17A mRNA expression and Th17-related transcription factors (RORγt and STAT3) in tissue samples. RESULTS The findings showed a substantial increase in Th17 cell percentages, elevated concentrations of IL-17A and IL-23, and higher mRNA expression levels of IL-17A, RORγt, and STAT3 in patients with PAC when compared to healthy controls (HCs). CONCLUSION Th17 cells play an important role in the pathogenesis of PAC and may represent potential therapeutic targets.
Collapse
Affiliation(s)
- Imteyaz Ahmad Khan
- Department of Gastroenterology and Human Nutrition Unit, All India Institute of Medical Sciences, New Delhi, India
| | - Nidhi Singh
- Department of Gastroenterology and Human Nutrition Unit, All India Institute of Medical Sciences, New Delhi, India
| | - Deepak Gunjan
- Department of Gastroenterology and Human Nutrition Unit, All India Institute of Medical Sciences, New Delhi, India
| | - Nihar Ranjan Dash
- Department of Gastrointestinal Surgery, All India Institute of Medical Sciences, New Delhi, India
| | - Baibaswata Nayak
- Department of Gastroenterology and Human Nutrition Unit, All India Institute of Medical Sciences, New Delhi, India
| | - Surabhi Gupta
- Department of Reproductive Biology, All India Institute of Medical Sciences, New Delhi, India
| | - Anoop Saraya
- Department of Gastroenterology and Human Nutrition Unit, All India Institute of Medical Sciences, New Delhi, India.
| |
Collapse
|
209
|
Jayathilake AG, Luwor RB, Nurgali K, Su XQ. Molecular Mechanisms Associated with the Inhibitory Role of Long Chain n-3 PUFA in Colorectal Cancer. Integr Cancer Ther 2024; 23:15347354241243024. [PMID: 38708673 PMCID: PMC11072084 DOI: 10.1177/15347354241243024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 02/14/2024] [Accepted: 03/11/2024] [Indexed: 05/07/2024] Open
Abstract
Colorectal cancer (CRC) is the third leading cause of cancer-related death in the world. Multiple evidence suggests that there is an association between excess fat consumption and the risk of CRC. The long chain n-3 polyunsaturated fatty acids (LC n-3 PUFA), especially eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA), are essential for human health, and both in vitro and in vivo studies have shown that these fatty acids can prevent CRC development through various molecular mechanisms. These include the modulation of arachidonic acid (AA) derived prostaglandin synthesis, alteration of growth signaling pathways, arrest of the cell cycle, induction of cell apoptosis, suppression of angiogenesis and modulation of inflammatory response. Human clinical studies found that LC n-3 PUFA combined with chemotherapeutic agents can improve the efficacy of treatment and reduce the dosage of chemotherapy and associated side effects. In this review, we discuss comprehensively the anti-cancer effects of LC n-3 PUFA on CRC, with a main focus on the underlying molecular mechanisms.
Collapse
Affiliation(s)
| | - Rodney Brain Luwor
- The University of Melbourne, Melbourne, VIC, Australia
- Fiona Elsey Cancer Research Institute, Ballarat, VIC, Australia
| | - Kulmira Nurgali
- Institute for Health and Sport, Victoria University, Melbourne, VIC, Australia
- The University of Melbourne, Melbourne, VIC, Australia
- Australian Institute for Muscular Skeletal Science (AIMSS), Melbourne, VIC, Australia
| | - Xiao Qun Su
- Institute for Health and Sport, Victoria University, Melbourne, VIC, Australia
| |
Collapse
|
210
|
Dantkale KS, Agrawal M. A Comprehensive Review of Current Trends in the Diagnosis and Treatment of Ovarian Germ Cell Tumors. Cureus 2024; 16:e52650. [PMID: 38380211 PMCID: PMC10877227 DOI: 10.7759/cureus.52650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Accepted: 01/20/2024] [Indexed: 02/22/2024] Open
Abstract
Ovarian germ cell tumors constitute a rare and intricate spectrum of neoplasms characterized by diverse histological subtypes. This comprehensive review elucidates the classification, diagnosis, treatment, prognosis, and unique challenges associated with these tumors. The classification is rooted in histological attributes, with principal subtypes encompassing dysgerminoma, immature teratoma, yolk sac tumor (endodermal sinus tumor), choriocarcinoma, and mixed germ cell tumors. Each subtype bears distinct characteristics and clinical implications, necessitating precise diagnosis and tailored therapeutic strategies. Diagnosis hinges upon recognizing the broad clinical presentation, employing imaging techniques (such as ultrasound and MRI), evaluating tumor markers (alpha-fetoprotein and beta-human chorionic gonadotropin), and conducting histopathological examinations where necessary. Staging, primarily utilizing the International Federation of Gynecology and Obstetrics (FIGO) system, is pivotal in determining the extent of disease, guiding treatment choices, and facilitating prognostic assessment. Treatment modalities encompass surgery, chemotherapy (including standard regimens and emerging therapies), radiation therapy, targeted therapies, and immunotherapy. Prognosis is influenced by histological subtype, tumor stage, patient age, surgical success, response to chemotherapy, and tumor markers, while predictive biomarkers are continually emerging. Despite advances in treatment, ovarian germ cell tumors pose distinct challenges, including late diagnosis, treatment-related side effects, and the enigma of chemoresistance. An integral aspect of comprehensive care is supportive strategies to manage symptoms and offer psychological and emotional support. This review accentuates the vital role of early diagnosis and multidisciplinary care in optimizing outcomes. Future research directions and evolving clinical practices are explored in these intricate and distinctive malignancies, highlighting the dynamic landscape of ovarian germ cell tumors.
Collapse
Affiliation(s)
- Ketki S Dantkale
- Department of Obstetrics and Gynecology, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education & Research (DMIHER), Wardha, IND
| | - Manjusha Agrawal
- Department of Obstetrics and Gynecology, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education & Research (DMIHER), Wardha, IND
| |
Collapse
|
211
|
Paly VF, Li S, Khanduri P, Asfaw AA, Zou D, Hernandez L. Budget impact analysis of introducing fruquintinib for metastatic colorectal cancer previously treated with fluoropyrimidine-, oxaliplatin-, and irinotecan-based chemotherapy and biologics in the United States from the payer perspective. J Med Econ 2024; 27:1076-1085. [PMID: 39102473 DOI: 10.1080/13696998.2024.2389005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 08/02/2024] [Accepted: 08/02/2024] [Indexed: 08/07/2024]
Abstract
AIMS Fruquintinib is a selective small molecule tyrosine kinase inhibitor of vascular endothelial growth factor receptor (VEGFR)-1, -2, and -3 recently approved in the United States (US) for the treatment of adult patients with metastatic colorectal cancer (CRC) who have previously been treated with fluoropyrimidine-, oxaliplatin-, and irinotecan-based chemotherapy, an anti-VEGF biological therapy, and if RAS wild-type and medically appropriate, anti-epidermal growth factor receptor therapy. This study aimed to estimate the 5-year budget impact of fruquintinib from a US payer perspective (commercial and Medicare). MATERIALS AND METHODS A budget impact model was developed to compare two scenarios: a reference scenario in which patients received regorafenib, trifluridine/tipiracil, or trifluridine/tipiracil with bevacizumab and an alternative scenario in which patients received reference scenario treatments or fruquintinib. Market shares were evenly divided across available options. A 5-year time horizon and a hypothetical health plan of 1 million members was assumed. The model included epidemiological inputs to estimate the eligible population; clinical inputs for treatment duration, progression-free survival, overall survival, and adverse event (AE) frequency; and cost inputs for treatment, AEs, disease management, subsequent therapy, and terminal care costs. Budget impact was reported as total, per member per year (PMPY), and per member per month (PMPM). RESULTS The model estimated an eligible population of 194 patients (39 per year) over 5 years. In the base case, the estimated 5-year budget impact of fruquintinib was $4,077,073 ($0.82 PMPY and 0.07 PMPM) for a commercial health plan. During the first year, the estimated budget impact was $627,570 ($0.63 PMPY and 0.05 PMPM). Results were robust across sensitivity analyses. PMPM costs from the Medicare perspective were greater than the base-case (commercial) ($0.17 vs. $0.07) due to higher incidence of CRC in that population. CONCLUSIONS Fruquintinib is associated with a low budget impact for payers based on proposed thresholds in the US.
Collapse
|
212
|
McCarthy G, Young K, Madin-Warburton M, Mantaian T, Brook E, Metcalfe K, Mikelson J, Xu R, Seyla-Hammer C, Aguiar-Ibáñez R, Amonkar M. Cost-effectiveness of pembrolizumab for previously treated MSI-H/dMMR solid tumours in the UK. J Med Econ 2024; 27:279-291. [PMID: 38293714 DOI: 10.1080/13696998.2024.2311507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 01/25/2024] [Indexed: 02/01/2024]
Abstract
OBJECTIVES Patients with previously treated microsatellite instability-high (MSI-H)/mismatch repair deficient (dMMR) tumours have limited chemotherapeutic treatment options. Pembrolizumab received approval from the EMA in 2022 for the treatment of colorectal, endometrial, gastric, small intestine, and biliary MSI-H/dMMR tumour types. This approval was supported by data from the KEYNOTE-164 and KEYNOTE-158 clinical trials. This study evaluated the cost-effectiveness of pembrolizumab compared with standard of care (SoC) for previously treated MSI-H/dMMR solid tumours in line with the approved EMA label from a UK healthcare payer perspective. METHODS A multi-tumour partitioned survival model was built consisting of pre-progression, progressed disease, and dead health states. Pembrolizumab survival outcomes were extrapolated using Bayesian hierarchical models (BHMs) fitted to pooled data from KEYNOTE-164 and KEYNOTE-158. Comparator outcomes were informed by published sources. Tumour sites were modelled independently and then combined, weighted by tumour site distribution. A SoC comparator was used to formulate the overall cost-effectiveness result with pembrolizumab as the intervention. SoC comprised a weighted average of the comparators by tumour site based on market share. Drug acquisition, administration, adverse events, monitoring, subsequent treatment, end-of-life costs, and testing costs were included. Sensitivity and scenario analyses were performed, including modelling pembrolizumab efficacy using standard parametric survival models. RESULTS Pembrolizumab, at list price, was associated with £129,469 in total costs, 8.30 LYs, and 3.88 QALYs across the pooled tumour sites. SoC was associated with £28,222 in total costs, 1.14 LYs, and 0.72 QALYs across the pooled tumour sites. This yields an incremental cost-effectiveness ratio (ICER) of £32,085 per QALY. Results were robust to sensitivity and scenario analyses. CONCLUSIONS This model demonstrates pembrolizumab provides a valuable new alternative therapy for UK patients with MSH-H/dMMR cancer at the cost of £32,085 per QALY, with confidential discounts anticipated to improve cost-effectiveness further.
Collapse
|
213
|
Gu Z, Wang L, Zhai MA J, Zhang T, Yang Y. Conventional Therapy Combined With Quxie Capsule Modulating Gut Microbiome in Metastatic Colorectal Cancer Patients With the Third or Above Line Setting: Result From an Investigator-Initiated, Open-Label, Single-Arm, Phase II Study. Integr Cancer Ther 2024; 23:15347354241280390. [PMID: 39313964 PMCID: PMC11425756 DOI: 10.1177/15347354241280390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 08/17/2024] [Accepted: 08/19/2024] [Indexed: 09/25/2024] Open
Abstract
BACKGROUND In patients with metastatic colorectal cancer (mCRC), Quxie Cap-sule (QX)-a combination of conventional therapy (including chemotherapy, targeted therapy or supportive care)-has shown a significant overall survival benefit compared with placebo and might have the property of dual effects of antitumor and immunity enhancement, both mediated by the microbiome. In preclinical models, QX has also shown activity against colorectal cancer. This study aimed to describe how the aforementioned effects of QX look after when focusing on the patients in third or above line setting. METHODS A Simon's Minimax two-stage phase II design was used in this study, which enrolled mCRC patients who progressed after second-line treatment. Patients received conventional therapy plus QX until disease progression or unacceptable toxicity. Before and after 1-month intervention, we collected patients' stool samples for microbiome analysis by 16s rRNA sequencing approaches. And the microbiome analysis before and after 1-month intervention was done through bioinformation analysis platform. RESULTS Fifteen patients were enrolled and gut microbiome were analyzed from 7 of 10 patients that with PFS over 3.7 months. Microbiome community analysis on genus level showed that the proportion of Lachnospiraceae_UCG-001 (0.04% vs 1.06%, P = .02249) significantly increased after conventional therapy plus QX while the proportion of Alistipes (2.96% vs 1.35%, P = .03461), Flavonifractor (0.04% vs 0.02%, P = .02249), Bifidobacterium (6.11% vs 1.14%, P = .02249) and Butyricimonas (0.24% vs 0.11%, P = .03603) significantly decreased after intervention . LEfSe analysis showed that after intervention, samples were highly related with unclassified-f-lachnospiraceae, Eubacterium and Lachnospiraceae_UCG-001. CONCLUSIONS Decrease of gut bacteria with potential roles in carcinogenesis of colorectal cancer and increase in the abundance of gut anticancer bacteria such as Lachnospiraceae may partly explain how conventional therapy combined with QX can influence carcinogenesis and tumor progression in colon cancer. TRIAL REGISTRATION Chinese Clinical Trial Registry (ChiCTR2100053874).
Collapse
Affiliation(s)
- Zhien Gu
- China Academy of Chinese Medical Sciences, Beijing, China
| | - Lei Wang
- Chongqing University Three Gorges Hospital, Chongqing, China
| | - Jiawei Zhai MA
- Beijing Chaoyang Integrative Medicinerescue and First Aid Hospital, Beijing, China
| | - Tong Zhang
- China Academy of Chinese Medical Sciences, Beijing, China
| | - Yufei Yang
- China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
214
|
Trulson I, Holdenrieder S. Prognostic value of blood-based protein biomarkers in non-small cell lung cancer: A critical review and 2008-2022 update. Tumour Biol 2024; 46:S111-S161. [PMID: 37927288 DOI: 10.3233/tub-230009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2023] Open
Abstract
BACKGROUND Therapeutic possibilities for non-small cell lung cancer (NSCLC) have considerably increased during recent decades. OBJECTIVE To summarize the prognostic relevance of serum tumor markers (STM) for early and late-stage NSCLC patients treated with classical chemotherapies, novel targeted and immune therapies. METHODS A PubMed database search was conducted for prognostic studies on carcinoembryonic antigen (CEA), cytokeratin-19 fragment (CYFRA 21-1), neuron-specific enolase, squamous-cell carcinoma antigen, progastrin-releasing-peptide, CA125, CA 19-9 and CA 15-3 STMs in NSCLC patients published from 2008 until June 2022. RESULTS Out of 1069 studies, 141 were identified as meeting the inclusion criteria. A considerable heterogeneity regarding design, patient number, analytical and statistical methods was observed. High pretherapeutic CYFRA 21-1 levels and insufficient decreases indicated unfavorable prognosis in many studies on NSCLC patients treated with chemo-, targeted and immunotherapies or their combinations in early and advanced stages. Similar results were seen for CEA in chemotherapy, however, high pretherapeutic levels were sometimes favorable in targeted therapies. CA125 is a promising prognostic marker in patients treated with immunotherapies. Combinations of STMs further increased the prognostic value over single markers. CONCLUSION Protein STMs, especially CYFRA 21-1, have prognostic potential in early and advanced stage NSCLC. For future STM investigations, better adherence to comparable study designs, analytical methods, outcome measures and statistical evaluation standards is recommended.
Collapse
Affiliation(s)
- Inga Trulson
- Munich Biomarker Research Center, Institute for Laboratory Medicine, German Heart Center, Technical University of Munich, Munich, Germany
| | - Stefan Holdenrieder
- Munich Biomarker Research Center, Institute for Laboratory Medicine, German Heart Center, Technical University of Munich, Munich, Germany
| |
Collapse
|
215
|
Tarpgaard LS, Winther SB, Pfeiffer P. Treatment Options in Late-Line Colorectal Cancer: Lessons Learned from Recent Randomized Studies. Cancers (Basel) 2023; 16:126. [PMID: 38201553 PMCID: PMC10777930 DOI: 10.3390/cancers16010126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 12/18/2023] [Accepted: 12/23/2023] [Indexed: 01/12/2024] Open
Abstract
Systemic treatment of metastatic colorectal cancer (mCRC) has improved considerably over the past 20 years. First- and second-line combinations of 5FU, oxaliplatin, and irinotecan, with or without anti-angiogenic and/or anti-EGFR antibodies, were approved shortly after the turn of the millennium. Further triumphs were not seen for almost 10 years, until the approval of initially regorafenib and shortly after trifluridine/tipiracil. A growing understanding of tumor biology through molecular profiling has led to further treatment options. Here, we review the most recent clinical data for late-line treatment options in mCRC, focusing on randomized trials if available. We include recommendations for options in unselected patients and therapies that should only be offered in patients with distinct tumor profiles (e.g., BRAF mutations, KRAS G12C mutations, HER2 amplification, deficient MMR, or NTRK gene fusions).
Collapse
Affiliation(s)
- Line Schmidt Tarpgaard
- Department of Oncology, Odense University Hospital, 5000 Odense C, Denmark; (S.B.W.); (P.P.)
- Department of Clinical Research, University of Southern Denmark, 5230 Odense M, Denmark
| | | | - Per Pfeiffer
- Department of Oncology, Odense University Hospital, 5000 Odense C, Denmark; (S.B.W.); (P.P.)
- Department of Clinical Research, University of Southern Denmark, 5230 Odense M, Denmark
| |
Collapse
|
216
|
Nakashima M, Li K, Chen Q, de Silva S, Li H, Kawakami K, Wei Q, Luo S, Zhao H. Appropriate dose of regorafenib based on body weight of colorectal cancer patients: a retrospective cohort study. BMC Cancer 2023; 23:1268. [PMID: 38129822 PMCID: PMC10740272 DOI: 10.1186/s12885-023-11720-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 12/05/2023] [Indexed: 12/23/2023] Open
Abstract
PURPOSE Previous randomized studies have shown a survival benefit of using regorafenib but a high rate of adverse events in unresectable colorectal cancer patients. To reduce these adverse events and improve the tolerability, we examined the appropriate dose of regorafenib based on body weight. METHODS We used a nationwide claims database in Japan and examined the efficacy and safety of regorafenib for patients with metastatic colorectal cancer between groups divided by body weight (60 kg) and median average dose (120 mg) between 2013 and 2018. We also assessed overall survival (OS) and adverse events between these groups. RESULTS We identified 2530 Japanese patients (heavy weight/high dose: 513, light weight/low dose: 921, heavy weight/low dose: 452, and light weight/high dose: 644). There was no significant difference in the adverse events and OS after inverse probability treatment weighting (IPTW) adjustment between heavy weight/high dose group and light weight/low dose group (hazard ratio, HR=0.97). Among the light-weight patients, higher average dose was associated with shorter OS (IPTW adjusted HR=1.21, 95% CI 1.05 - 1.39, Table 3) while among the heavy-weight patients, there was no significant difference in OS between high and low dose groups (IPTW adjusted HR=1.14, 95% CI 0.95 - 1.37). CONCLUSION The findings suggest that a low dose of regorafenib for light-weight patients may be as safe and effective as high doses for heavy-weight patients. Further studies should be conducted to identify an appropriate dose based on each patient's physique and condition.
Collapse
Affiliation(s)
- Masayuki Nakashima
- Department of Pharmacoepidemiology, Graduate School of Medicine and Public Health, Kyoto University, Kyoto, Japan
| | - Kan Li
- MRL, Merck & Co., Inc, North Wales, PA, USA
| | - Qichen Chen
- Department of Hepatobiliary Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | | | - Hal Li
- MRL, Merck & Co., Inc, North Wales, PA, USA
| | - Koji Kawakami
- Department of Pharmacoepidemiology, Graduate School of Medicine and Public Health, Kyoto University, Kyoto, Japan
| | - Qingyi Wei
- Duke Cancer Institute, Duke University Medical Center, Durham, NC, USA
- Department of Medicine, Duke University School of Medicine, Durham, NC, USA
- Department of Population Health Sciences, Duke University, Durham, NC, USA
| | - Sheng Luo
- Department of Biostatistics and Bioinformatics, Duke University, Durham, NC, USA.
| | - Hong Zhao
- Department of Hepatobiliary Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
217
|
Davis CH, Laird AM, Libutti SK. Resistant gastroenteropancreatic neuroendocrine tumors: a definition and guideline to medical and surgical management. Proc AMIA Symp 2023; 37:104-110. [PMID: 38174011 PMCID: PMC10761146 DOI: 10.1080/08998280.2023.2284039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 11/07/2023] [Indexed: 01/05/2024] Open
Abstract
Gastroenteropancreatic neuroendocrine tumors (NETs), also historically known as carcinoids, are tumors derived of hormone-secreting enteroendocrine cells. Carcinoids may be found in the esophagus, stomach, small intestine, appendix, colon, rectum, or pancreas. The biologic behavior of carcinoids differs based on their location, with gastric and appendiceal NETs among the least aggressive and small intestinal and pancreatic NETs among the most aggressive. Ultimately, however, biologic behavior is most heavily influenced by tumor grade. The incidence of NETs has increased by 6.4 times over the past 40 years. Surgery remains the mainstay for management of most carcinoids. Medical management, however, is a useful adjunct and/or definitive therapy in patients with symptomatic functional carcinoids, in patients with unresectable or incompletely resected carcinoids, in some cases of recurrent carcinoid, and in postoperative patients to prevent recurrence. Functional tumors with persistent symptoms or progressive metastatic carcinoids despite therapy are called "resistant" tumors. In patients with unresectable disease and/or carcinoid syndrome, an array of medical therapies is available, mainly including somatostatin analogues, molecular-targeted therapy, and peptide receptor radionuclide therapy. Active research is ongoing to identify additional targeted therapies for patients with resistant carcinoids.
Collapse
Affiliation(s)
- Catherine H. Davis
- Division of Surgical Oncology, Baylor University Medical Center, Dallas, Texas, USA
- Texas A&M University School of Medicine, Dallas, Texas, USA
| | - Amanda M. Laird
- Division of Surgical Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey, USA
- Rutgers Robert Wood Johnson University Medical School, New Brunswick, New Jersey, USA
| | - Steven K. Libutti
- Division of Surgical Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey, USA
- Rutgers Robert Wood Johnson University Medical School, New Brunswick, New Jersey, USA
| |
Collapse
|
218
|
Gambaro K, Marques M, McNamara S, Couetoux du Tertre M, Hoffert C, Srivastava A, Schab A, Alcindor T, Langleben A, Sideris L, Abdelsalam M, Tehfe M, Couture F, Batist G, Kavan P. A Phase II Exploratory Study to Identify Biomarkers Predictive of Clinical Response to Regorafenib in Patients with Metastatic Colorectal Cancer Who Have Failed First-Line Therapy. Int J Mol Sci 2023; 25:43. [PMID: 38203214 PMCID: PMC10778949 DOI: 10.3390/ijms25010043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 12/15/2023] [Accepted: 12/16/2023] [Indexed: 01/12/2024] Open
Abstract
Single-agent regorafenib is approved in Canada for metastatic colorectal cancer (mCRC) patients who have failed previous lines of therapy. Identifying prognostic biomarkers is key to optimizing therapeutic strategies for these patients. In this clinical study (NCT01949194), we evaluated the safety and efficacy of single-agent regorafenib as a second-line therapy for mCRC patients who received it after failing first-line therapy with an oxaliplatin or irinotecan regimen with or without bevacizumab. Using various omics approaches, we also investigated putative biomarkers of response and resistance to regorafenib in metastatic lesions and blood samples in the same cohort. Overall, the safety profile of regorafenib seemed similar to the CORRECT trial, where regorafenib was administered as ≥ 2 lines of therapy. While the mutational landscape showed typical mutation rates for the top five driver genes (APC, KRAS, BRAF, PIK3CA, and TP53), KRAS mutations were enriched in intrinsically resistant lesions. Additional exploration of genomic-phenotype associations revealed several biomarker candidates linked to unfavorable prognoses in patients with mCRC using various approaches, including pathway analysis, cfDNA profiling, and copy number analysis. However, further research endeavors are necessary to validate the potential utility of these promising genes in understanding patients' responses to regorafenib treatment.
Collapse
Affiliation(s)
- Karen Gambaro
- Canadian National Centres of Excellence-Exactis Innovations, Montreal, QC H3T 1Y6, Canada; (K.G.); (M.M.); (S.M.)
- Consortium de Recherche en Oncologie Clinique du Québec (Q-CROC), Quebec, QC G1V 3X8, Canada
- Segal Cancer Centre-Jewish General Hospital, Montreal, QC H3T 1E2, Canada
| | - Maud Marques
- Canadian National Centres of Excellence-Exactis Innovations, Montreal, QC H3T 1Y6, Canada; (K.G.); (M.M.); (S.M.)
- Segal Cancer Centre-Jewish General Hospital, Montreal, QC H3T 1E2, Canada
| | - Suzan McNamara
- Canadian National Centres of Excellence-Exactis Innovations, Montreal, QC H3T 1Y6, Canada; (K.G.); (M.M.); (S.M.)
- Consortium de Recherche en Oncologie Clinique du Québec (Q-CROC), Quebec, QC G1V 3X8, Canada
| | - Mathilde Couetoux du Tertre
- Canadian National Centres of Excellence-Exactis Innovations, Montreal, QC H3T 1Y6, Canada; (K.G.); (M.M.); (S.M.)
- Consortium de Recherche en Oncologie Clinique du Québec (Q-CROC), Quebec, QC G1V 3X8, Canada
| | - Cyrla Hoffert
- Canadian National Centres of Excellence-Exactis Innovations, Montreal, QC H3T 1Y6, Canada; (K.G.); (M.M.); (S.M.)
- Consortium de Recherche en Oncologie Clinique du Québec (Q-CROC), Quebec, QC G1V 3X8, Canada
- Segal Cancer Centre-Jewish General Hospital, Montreal, QC H3T 1E2, Canada
| | - Archana Srivastava
- Canadian National Centres of Excellence-Exactis Innovations, Montreal, QC H3T 1Y6, Canada; (K.G.); (M.M.); (S.M.)
- Consortium de Recherche en Oncologie Clinique du Québec (Q-CROC), Quebec, QC G1V 3X8, Canada
- Segal Cancer Centre-Jewish General Hospital, Montreal, QC H3T 1E2, Canada
| | - Anna Schab
- Canadian National Centres of Excellence-Exactis Innovations, Montreal, QC H3T 1Y6, Canada; (K.G.); (M.M.); (S.M.)
- Consortium de Recherche en Oncologie Clinique du Québec (Q-CROC), Quebec, QC G1V 3X8, Canada
| | | | | | - Lucas Sideris
- Hôpital Maisonneuve Rosemont, Montreal, QC H1T 2M4, Canada
| | | | - Mustapha Tehfe
- Hematology-Oncology, Oncology Center-Centre Hospitalier de l’Université de Montreal, Montreal, QC H2X 0C1, Canada
| | | | - Gerald Batist
- Segal Cancer Centre-Jewish General Hospital, Montreal, QC H3T 1E2, Canada
| | - Petr Kavan
- Segal Cancer Centre-Jewish General Hospital, Montreal, QC H3T 1E2, Canada
| |
Collapse
|
219
|
Ozkan M, Oflazoglu U, Yildiz Y, Güc ZG, Salman T, Ünal S, Kücükzeybek Y, Alacacioglu A. Regorafenib therapy as a third-line treatment for metastatic colorectal cancer: A single center long term experience. Medicine (Baltimore) 2023; 102:e36435. [PMID: 38115370 PMCID: PMC10727636 DOI: 10.1097/md.0000000000036435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 11/01/2023] [Accepted: 11/10/2023] [Indexed: 12/21/2023] Open
Abstract
This study examined the effects of regorafenib (Reg) on progression-free survival (PFS), overall survival (OS), and adverse events (AEs) in metastatic colorectal cancer (mCRC) patients who underwent targeted treatment and chemotherapy. Reg was administered as a third-line treatment to 84 patients who had undergone 2 rounds of chemotherapy and targeted therapy and subsequently experienced progression. Treatment was initiated with a daily dose of 80 or 120 mg, based on the patient's ability to tolerate the medication, which was increased to 160 mg/day. The median PFS with Reg was 4 ± 0.2 months, while the median OS was 9 ± 1.2 months. When compared to patients who started Reg treatment at 80 mg, patients starting at 160 mg had longer median PFS and OS (PFS:6 ± 2.1 months vs 4 ± 0.2 months; P = .05; OS:13 ± 0.7 months vs 6 ± 1.3 months; P = .069). Patients with right-sided colon cancer who received Reg as third-line therapy had a significantly longer mPFS than those with left-sided colon cancer (8 months ± 4 vs 4 months ± 0.3, P = .039). Patients with KRAS mutations had a prolonged mPFS than those with panRAS-wild type (6 ± 1.6 months vs 4 ± 0.3 months, P = .06). The mPFS contribution in the BRAF mutant subgroup with poor prognosis is promising, as it is similar to that of patients without BRAF mutations (4 months ± 0.8 × 4 months ± 0.5, P = .74). The most common AEs reported were elevated liver enzyme levels and dermatological toxicities.
Collapse
Affiliation(s)
- Merve Ozkan
- İzmir Katip Celebi University, Department of Medical Oncology, Izmir, Turkey
| | - Utku Oflazoglu
- İzmir Katip Celebi University, Department of Medical Oncology, Izmir, Turkey
| | - Yaşar Yildiz
- İzmir Katip Celebi University, Department of Medical Oncology, Izmir, Turkey
| | - Zeynep G. Güc
- İzmir Katip Celebi University, Department of Medical Oncology, Izmir, Turkey
| | - Tarik Salman
- İzmir Katip Celebi University, Department of Medical Oncology, Izmir, Turkey
| | - Sinan Ünal
- İzmir Katip Celebi University, Department of Medical Oncology, Izmir, Turkey
| | - Yüksel Kücükzeybek
- İzmir Katip Celebi University, Department of Medical Oncology, Izmir, Turkey
| | - Ahmet Alacacioglu
- İzmir Katip Celebi University, Department of Medical Oncology, Izmir, Turkey
| |
Collapse
|
220
|
Puttagunta P, Pamulapati SV, Bates JE, Gross JH, Stokes WA, Schmitt NC, Steuer C, Teng Y, Saba NF. Critical review of the current and future prospects of VEGF-TKIs in the management of squamous cell carcinoma of head and neck. Front Oncol 2023; 13:1310106. [PMID: 38192624 PMCID: PMC10773827 DOI: 10.3389/fonc.2023.1310106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 11/28/2023] [Indexed: 01/10/2024] Open
Abstract
As the prognosis for squamous cell carcinoma of the head and neck remains unsatisfactory when compared to other malignancies, novel therapies targeting specific biomarkers are a critical emerging area of great promise. One particular class of drugs that has been developed to impede tumor angiogenesis is vascular endothelial growth factor-tyrosine kinase inhibitors. As current data is primarily limited to preclinical and phase I/II trials, this review summarizes the current and future prospects of these agents in squamous cell carcinoma of the head and neck. In particular, the combination of these agents with immunotherapy is an exciting area that may be a promising option for patients with recurrent or metastatic disease, evidenced in recent trials such as the combination immune checkpoint inhibitors with lenvatinib and cabozantinib. In addition, the use of such combination therapy preoperatively in locally advanced disease is another area of interest.
Collapse
Affiliation(s)
- Prashant Puttagunta
- Medical Education, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Saagar V. Pamulapati
- Internal Medicine Program, Mercyhealth Graduate Medical Education Consortium, Rockford, IL, United States
| | - James E. Bates
- Department of Radiation Oncology, Emory University School of Medicine, Atlanta, GA, United States
| | - Jennifer H. Gross
- Department of Otolaryngology – Head and Neck Surgery, Emory University School of Medicine, Atlanta, GA, United States
| | - William A. Stokes
- Department of Radiation Oncology, Emory University School of Medicine, Atlanta, GA, United States
| | - Nicole C. Schmitt
- Department of Otolaryngology – Head and Neck Surgery, Emory University School of Medicine, Atlanta, GA, United States
| | - Conor Steuer
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, United States
| | - Yong Teng
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, United States
| | - Nabil F. Saba
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, United States
| |
Collapse
|
221
|
Passardi A, Gibbons D. Editorial: Molecular targets for the treatment of metastatic colorectal cancer. Front Oncol 2023; 13:1341594. [PMID: 38156115 PMCID: PMC10752799 DOI: 10.3389/fonc.2023.1341594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 11/21/2023] [Indexed: 12/30/2023] Open
Affiliation(s)
- Alessandro Passardi
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - David Gibbons
- Department of Pathology, St Vincent’s University Hospital, Dublin, Ireland
- School of Medicine and Medical Sciences, University College Dublin, Dublin, Ireland
| |
Collapse
|
222
|
Signorelli C, Chilelli MG, Giannarelli D, Basso M, Calegari MA, Anghelone A, Lucchetti J, Minelli A, Angotti L, Zurlo IV, Schirripa M, Morelli C, Dell’Aquila E, Cosimati A, Gemma D, Ribelli M, Emiliani A, Corsi DC, Arrivi G, Mazzuca F, Zoratto F, Morandi MG, Santamaria F, Saltarelli R, Ruggeri EM. Retrospective Correlation between First Drug Treatment Duration and Survival Outcomes in Sequential Treatment with Regorafenib and Trifluridine/Tipiracil in Refractory Metastatic Colorectal Cancer: A Real-World Subgroup Analysis. Cancers (Basel) 2023; 15:5758. [PMID: 38136304 PMCID: PMC10741389 DOI: 10.3390/cancers15245758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 12/03/2023] [Accepted: 12/06/2023] [Indexed: 12/24/2023] Open
Abstract
Background: Patients with refractory metastatic colorectal cancer (mCRC) rarely receive third-line or further treatment. In this context, regorafenib (R) and trifluridine/tipiracil (T) are two important novel therapeutic choices with statistically significant increases in overall survival (OS), progression-free survival (PFS), and disease control, with different toxicity profiles. This study is a subgroup analysis of our larger retrospective study, already published, whose objective was to assess the outcomes of patients when R and T were given sequentially. Patients and Methods: The study involved thirteen Italian cancer centers on a 10-year retrospective observation (2012-2022). In this subgroup analysis, we focused our attention on the correlation between the first drug treatment duration (<3 months, 3 to <6 months and ≥6 months) and survival outcomes in patients who had received the sequence regorafenib-to-trifluridine/tipiracil, or vice versa. Results: The initial study included 866 patients with mCRC who received sequential T/R, or R/T, or T or R alone. This analysis is focused on evaluating the impact of the duration of the first treatment in the sequence on clinical outcomes (OS, PFS) and includes 146 and 116 patients of the T/R and R/T sequences, respectively. Based on the duration of the first drug treatment, subgroups for the T/R sequence included 27 patients (18.4%) who received T for <3 months, 86 (58.9%) treated for 3 to <6 months, and 33 (22.6%) treated for ≥6 months; in the reverse sequence (R as the first drug), subgroups included 18 patients (15.5%) who received their first treatment for <3 months, 62 (53.4%) treated for 3 to <6 months, and 35 (31.0%) treated for ≥6 months. In patients who received their first drug treatment for a period of 3 to <6 months, the R/T sequence had a significantly longer median OS (13.7 vs. 10.8 months, p = 0.0069) and a longer median PFS (10.8 vs. 8.5 months, p = 0.0003) than the T/R group. There were no statistically significant differences between groups with first drug treatment durations of <3 months and ≥6 months. Conclusions: Our analysis seems to suggest that the administration of R for a period of 3 to <6 months before that of T can prolong both OS and PFS, as compared to the opposite sequence.
Collapse
Affiliation(s)
- Carlo Signorelli
- Medical Oncology Unit, Belcolle Hospital, ASL Viterbo, 01100 Viterbo, Italy
| | | | - Diana Giannarelli
- Biostatistics Unit, Scientific Directorate, Fondazione Policlinico Universitario Agostino Gemelli, IRCCS, 00168 Rome, Italy
| | - Michele Basso
- Unit of Medical Oncology, Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli, IRCCS, 00168 Rome, Italy
| | - Maria Alessandra Calegari
- Unit of Medical Oncology, Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli, IRCCS, 00168 Rome, Italy
| | - Annunziato Anghelone
- Unit of Medical Oncology, Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli, IRCCS, 00168 Rome, Italy
| | - Jessica Lucchetti
- Division of Medical Oncology, Policlinico Universitario Campus Bio-Medico, 00128 Rome, Italy
| | - Alessandro Minelli
- Division of Medical Oncology, Policlinico Universitario Campus Bio-Medico, 00128 Rome, Italy
| | - Lorenzo Angotti
- Division of Medical Oncology, Policlinico Universitario Campus Bio-Medico, 00128 Rome, Italy
| | | | - Marta Schirripa
- Medical Oncology Unit, Belcolle Hospital, ASL Viterbo, 01100 Viterbo, Italy
| | - Cristina Morelli
- Medical Oncology Unit, Department of Systems Medicine, Tor Vergata University Hospital, 00133 Rome, Italy
| | - Emanuela Dell’Aquila
- Medical Oncology 1, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy
| | - Antonella Cosimati
- Medical Oncology 1, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy
| | - Donatello Gemma
- Medical Oncology Unit, ASL Frosinone, 03039 Sora (FR), Italy
| | - Marta Ribelli
- Medical Oncology Unit, Isola Tiberina Hospital-Gemelli Isola, 00186 Rome, Italy
| | - Alessandra Emiliani
- Medical Oncology Unit, Isola Tiberina Hospital-Gemelli Isola, 00186 Rome, Italy
| | | | - Giulia Arrivi
- Department of Clinical and Molecular Medicine, Oncology Unit, Sant’ Andrea University Hospital, Sapienza University of Rome, 00189 Rome, Italy
| | - Federica Mazzuca
- Department of Clinical and Molecular Medicine, Oncology Unit, Sant’ Andrea University Hospital, Sapienza University of Rome, 00189 Rome, Italy
| | | | - Maria Grazia Morandi
- Medical Oncology Unit, San Camillo de Lellis Hospital, ASL Rieti, 02100 Rieti, Italy
| | - Fiorenza Santamaria
- UOC Oncology A, Policlinico Umberto I, 00185 Rome, Italy
- Experimental Medicine, Network Oncology and Precision Medicine, Department of Experimental Medicine, Sapienza University of Rome, 00189 Rome, Italy
| | - Rosa Saltarelli
- UOC Oncology, San Giovanni Evangelista Hospital, ASL RM5, 00019 Tivoli (RM), Italy
| | - Enzo Maria Ruggeri
- Medical Oncology Unit, Belcolle Hospital, ASL Viterbo, 01100 Viterbo, Italy
| |
Collapse
|
223
|
Ottaiano A, Santorsola M. Revolutionizing KRAS p.G12C therapy in metastatic colorectal cancer: The triumph of dual inhibition. MED 2023; 4:857-859. [PMID: 38070479 DOI: 10.1016/j.medj.2023.11.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 11/09/2023] [Accepted: 11/10/2023] [Indexed: 12/18/2023]
Abstract
The management of refractory metastatic colorectal cancer patients with the KRAS p.G12C mutation presents a significant unmet need, with limited success using standard therapies. The study by Fakih et al. highlights the potential of sotorasib and panitumumab combination therapy in this clinical context, paving the way for a promising personalized therapeutic approach.1.
Collapse
Affiliation(s)
- Alessandro Ottaiano
- Department of Abdominal Oncology, Istituto Nazionale Tumori di Napoli, IRCCS "G. Pascale", via Mariano Semmola, 80131 Naples, Italy.
| | - Mariachiara Santorsola
- Department of Abdominal Oncology, Istituto Nazionale Tumori di Napoli, IRCCS "G. Pascale", via Mariano Semmola, 80131 Naples, Italy
| |
Collapse
|
224
|
Fakih MG, Salvatore L, Esaki T, Modest DP, Lopez-Bravo DP, Taieb J, Karamouzis MV, Ruiz-Garcia E, Kim TW, Kuboki Y, Meriggi F, Cunningham D, Yeh KH, Chan E, Chao J, Saportas Y, Tran Q, Cremolini C, Pietrantonio F. Sotorasib plus Panitumumab in Refractory Colorectal Cancer with Mutated KRAS G12C. N Engl J Med 2023; 389:2125-2139. [PMID: 37870968 DOI: 10.1056/nejmoa2308795] [Citation(s) in RCA: 70] [Impact Index Per Article: 70.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
BACKGROUND KRAS G12C is a mutation that occurs in approximately 3 to 4% of patients with metastatic colorectal cancer. Monotherapy with KRAS G12C inhibitors has yielded only modest efficacy. Combining the KRAS G12C inhibitor sotorasib with panitumumab, an epidermal growth factor receptor (EGFR) inhibitor, may be an effective strategy. METHODS In this phase 3, multicenter, open-label, randomized trial, we assigned patients with chemorefractory metastatic colorectal cancer with mutated KRAS G12C who had not received previous treatment with a KRAS G12C inhibitor to receive sotorasib at a dose of 960 mg once daily plus panitumumab (53 patients), sotorasib at a dose of 240 mg once daily plus panitumumab (53 patients), or the investigator's choice of trifluridine-tipiracil or regorafenib (standard care; 54 patients). The primary end point was progression-free survival as assessed by blinded independent central review according to the Response Evaluation Criteria in Solid Tumors, version 1.1. Key secondary end points were overall survival and objective response. RESULTS After a median follow-up of 7.8 months (range, 0.1 to 13.9), the median progression-free survival was 5.6 months (95% confidence interval [CI], 4.2 to 6.3) and 3.9 months (95% CI, 3.7 to 5.8) in the 960-mg sotorasib-panitumumab and 240-mg sotorasib-panitumumab groups, respectively, as compared with 2.2 months (95% CI, 1.9 to 3.9) in the standard-care group. The hazard ratio for disease progression or death in the 960-mg sotorasib-panitumumab group as compared with the standard-care group was 0.49 (95% CI, 0.30 to 0.80; P = 0.006), and the hazard ratio in the 240-mg sotorasib-panitumumab group was 0.58 (95% CI, 0.36 to 0.93; P = 0.03). Overall survival data are maturing. The objective response was 26.4% (95% CI, 15.3 to 40.3), 5.7% (95% CI, 1.2 to 15.7), and 0% (95% CI, 0.0 to 6.6) in the 960-mg sotorasib-panitumumab, 240-mg sotorasib-panitumumab, and standard-care groups, respectively. Treatment-related adverse events of grade 3 or higher occurred in 35.8%, 30.2%, and 43.1% of patients, respectively. Skin-related toxic effects and hypomagnesemia were the most common adverse events observed with sotorasib-panitumumab. CONCLUSIONS In this phase 3 trial of a KRAS G12C inhibitor plus an EGFR inhibitor in patients with chemorefractory metastatic colorectal cancer, both doses of sotorasib in combination with panitumumab resulted in longer progression-free survival than standard treatment. Toxic effects were as expected for either agent alone and resulted in few discontinuations of treatment. (Funded by Amgen; CodeBreaK 300 ClinicalTrials.gov number, NCT05198934.).
Collapse
Affiliation(s)
- Marwan G Fakih
- From Medical Oncology and Therapeutics Research, City of Hope Comprehensive Cancer Center, Duarte (M.G.F.), and Amgen, Thousand Oaks (E.C., J.C., Y.S., Q.T.) - both in California; Oncologia Medica, Università Cattolica del Sacro Cuore, and Oncologia Medica, Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome (L.S.), the Oncology Department, Fondazione Poliambulanza Istituto Ospedaliero, Brescia (F.M.), the Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa (C.C.), and the Medical Oncology Department, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico, Istituto Nazionale dei Tumori, Milan (F.P.) - all in Italy; the Department of Gastrointestinal and Medical Oncology, National Hospital Organization Kyushu Cancer Center, Fukuoka (T.E.), and the Experimental Therapeutics and GI Oncology Department, National Cancer Center Hospital East, Kashiwa (Y.K.) - both in Japan; the Medicine Department of Hematology, Oncology, and Tumor Immunology, Charité-Universitätsmedizin Berlin, Berlin (D.P.M.); the Department of Medical Oncology, Hospital de la Santa Creu i Sant Pau, Barcelona (D.P.L.-B.); Université Paris Cité, Site de Recherche Intégrée sur le Cancer, Cancer Research for Personalized Medicine Comprehensive Cancer Center, Department of Gastroenterology and Digestive Oncology, Hôpital Européen Georges-Pompidou, Paris (J.T.); the Department of Biological Chemistry, National and Kapodistrian University of Athens-School of Medicine, Athens (M.V.K.); Gastrointestinal Oncology Department and Translational Medicine Laboratory, Instituto Nacional de Cancerologia, Mexico City (E.R.-G.); the Oncology Department, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea (T.W.K.); the Medicine Department, Royal Marsden Hospital, London (D.C.); and the Department of Oncology, National Taiwan University Hospital, and the Graduate Institute of Oncology, National Taiwan University College of Medicine - both in Taipei (K.H.Y.)
| | - Lisa Salvatore
- From Medical Oncology and Therapeutics Research, City of Hope Comprehensive Cancer Center, Duarte (M.G.F.), and Amgen, Thousand Oaks (E.C., J.C., Y.S., Q.T.) - both in California; Oncologia Medica, Università Cattolica del Sacro Cuore, and Oncologia Medica, Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome (L.S.), the Oncology Department, Fondazione Poliambulanza Istituto Ospedaliero, Brescia (F.M.), the Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa (C.C.), and the Medical Oncology Department, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico, Istituto Nazionale dei Tumori, Milan (F.P.) - all in Italy; the Department of Gastrointestinal and Medical Oncology, National Hospital Organization Kyushu Cancer Center, Fukuoka (T.E.), and the Experimental Therapeutics and GI Oncology Department, National Cancer Center Hospital East, Kashiwa (Y.K.) - both in Japan; the Medicine Department of Hematology, Oncology, and Tumor Immunology, Charité-Universitätsmedizin Berlin, Berlin (D.P.M.); the Department of Medical Oncology, Hospital de la Santa Creu i Sant Pau, Barcelona (D.P.L.-B.); Université Paris Cité, Site de Recherche Intégrée sur le Cancer, Cancer Research for Personalized Medicine Comprehensive Cancer Center, Department of Gastroenterology and Digestive Oncology, Hôpital Européen Georges-Pompidou, Paris (J.T.); the Department of Biological Chemistry, National and Kapodistrian University of Athens-School of Medicine, Athens (M.V.K.); Gastrointestinal Oncology Department and Translational Medicine Laboratory, Instituto Nacional de Cancerologia, Mexico City (E.R.-G.); the Oncology Department, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea (T.W.K.); the Medicine Department, Royal Marsden Hospital, London (D.C.); and the Department of Oncology, National Taiwan University Hospital, and the Graduate Institute of Oncology, National Taiwan University College of Medicine - both in Taipei (K.H.Y.)
| | - Taito Esaki
- From Medical Oncology and Therapeutics Research, City of Hope Comprehensive Cancer Center, Duarte (M.G.F.), and Amgen, Thousand Oaks (E.C., J.C., Y.S., Q.T.) - both in California; Oncologia Medica, Università Cattolica del Sacro Cuore, and Oncologia Medica, Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome (L.S.), the Oncology Department, Fondazione Poliambulanza Istituto Ospedaliero, Brescia (F.M.), the Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa (C.C.), and the Medical Oncology Department, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico, Istituto Nazionale dei Tumori, Milan (F.P.) - all in Italy; the Department of Gastrointestinal and Medical Oncology, National Hospital Organization Kyushu Cancer Center, Fukuoka (T.E.), and the Experimental Therapeutics and GI Oncology Department, National Cancer Center Hospital East, Kashiwa (Y.K.) - both in Japan; the Medicine Department of Hematology, Oncology, and Tumor Immunology, Charité-Universitätsmedizin Berlin, Berlin (D.P.M.); the Department of Medical Oncology, Hospital de la Santa Creu i Sant Pau, Barcelona (D.P.L.-B.); Université Paris Cité, Site de Recherche Intégrée sur le Cancer, Cancer Research for Personalized Medicine Comprehensive Cancer Center, Department of Gastroenterology and Digestive Oncology, Hôpital Européen Georges-Pompidou, Paris (J.T.); the Department of Biological Chemistry, National and Kapodistrian University of Athens-School of Medicine, Athens (M.V.K.); Gastrointestinal Oncology Department and Translational Medicine Laboratory, Instituto Nacional de Cancerologia, Mexico City (E.R.-G.); the Oncology Department, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea (T.W.K.); the Medicine Department, Royal Marsden Hospital, London (D.C.); and the Department of Oncology, National Taiwan University Hospital, and the Graduate Institute of Oncology, National Taiwan University College of Medicine - both in Taipei (K.H.Y.)
| | - Dominik P Modest
- From Medical Oncology and Therapeutics Research, City of Hope Comprehensive Cancer Center, Duarte (M.G.F.), and Amgen, Thousand Oaks (E.C., J.C., Y.S., Q.T.) - both in California; Oncologia Medica, Università Cattolica del Sacro Cuore, and Oncologia Medica, Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome (L.S.), the Oncology Department, Fondazione Poliambulanza Istituto Ospedaliero, Brescia (F.M.), the Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa (C.C.), and the Medical Oncology Department, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico, Istituto Nazionale dei Tumori, Milan (F.P.) - all in Italy; the Department of Gastrointestinal and Medical Oncology, National Hospital Organization Kyushu Cancer Center, Fukuoka (T.E.), and the Experimental Therapeutics and GI Oncology Department, National Cancer Center Hospital East, Kashiwa (Y.K.) - both in Japan; the Medicine Department of Hematology, Oncology, and Tumor Immunology, Charité-Universitätsmedizin Berlin, Berlin (D.P.M.); the Department of Medical Oncology, Hospital de la Santa Creu i Sant Pau, Barcelona (D.P.L.-B.); Université Paris Cité, Site de Recherche Intégrée sur le Cancer, Cancer Research for Personalized Medicine Comprehensive Cancer Center, Department of Gastroenterology and Digestive Oncology, Hôpital Européen Georges-Pompidou, Paris (J.T.); the Department of Biological Chemistry, National and Kapodistrian University of Athens-School of Medicine, Athens (M.V.K.); Gastrointestinal Oncology Department and Translational Medicine Laboratory, Instituto Nacional de Cancerologia, Mexico City (E.R.-G.); the Oncology Department, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea (T.W.K.); the Medicine Department, Royal Marsden Hospital, London (D.C.); and the Department of Oncology, National Taiwan University Hospital, and the Graduate Institute of Oncology, National Taiwan University College of Medicine - both in Taipei (K.H.Y.)
| | - David P Lopez-Bravo
- From Medical Oncology and Therapeutics Research, City of Hope Comprehensive Cancer Center, Duarte (M.G.F.), and Amgen, Thousand Oaks (E.C., J.C., Y.S., Q.T.) - both in California; Oncologia Medica, Università Cattolica del Sacro Cuore, and Oncologia Medica, Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome (L.S.), the Oncology Department, Fondazione Poliambulanza Istituto Ospedaliero, Brescia (F.M.), the Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa (C.C.), and the Medical Oncology Department, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico, Istituto Nazionale dei Tumori, Milan (F.P.) - all in Italy; the Department of Gastrointestinal and Medical Oncology, National Hospital Organization Kyushu Cancer Center, Fukuoka (T.E.), and the Experimental Therapeutics and GI Oncology Department, National Cancer Center Hospital East, Kashiwa (Y.K.) - both in Japan; the Medicine Department of Hematology, Oncology, and Tumor Immunology, Charité-Universitätsmedizin Berlin, Berlin (D.P.M.); the Department of Medical Oncology, Hospital de la Santa Creu i Sant Pau, Barcelona (D.P.L.-B.); Université Paris Cité, Site de Recherche Intégrée sur le Cancer, Cancer Research for Personalized Medicine Comprehensive Cancer Center, Department of Gastroenterology and Digestive Oncology, Hôpital Européen Georges-Pompidou, Paris (J.T.); the Department of Biological Chemistry, National and Kapodistrian University of Athens-School of Medicine, Athens (M.V.K.); Gastrointestinal Oncology Department and Translational Medicine Laboratory, Instituto Nacional de Cancerologia, Mexico City (E.R.-G.); the Oncology Department, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea (T.W.K.); the Medicine Department, Royal Marsden Hospital, London (D.C.); and the Department of Oncology, National Taiwan University Hospital, and the Graduate Institute of Oncology, National Taiwan University College of Medicine - both in Taipei (K.H.Y.)
| | - Julien Taieb
- From Medical Oncology and Therapeutics Research, City of Hope Comprehensive Cancer Center, Duarte (M.G.F.), and Amgen, Thousand Oaks (E.C., J.C., Y.S., Q.T.) - both in California; Oncologia Medica, Università Cattolica del Sacro Cuore, and Oncologia Medica, Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome (L.S.), the Oncology Department, Fondazione Poliambulanza Istituto Ospedaliero, Brescia (F.M.), the Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa (C.C.), and the Medical Oncology Department, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico, Istituto Nazionale dei Tumori, Milan (F.P.) - all in Italy; the Department of Gastrointestinal and Medical Oncology, National Hospital Organization Kyushu Cancer Center, Fukuoka (T.E.), and the Experimental Therapeutics and GI Oncology Department, National Cancer Center Hospital East, Kashiwa (Y.K.) - both in Japan; the Medicine Department of Hematology, Oncology, and Tumor Immunology, Charité-Universitätsmedizin Berlin, Berlin (D.P.M.); the Department of Medical Oncology, Hospital de la Santa Creu i Sant Pau, Barcelona (D.P.L.-B.); Université Paris Cité, Site de Recherche Intégrée sur le Cancer, Cancer Research for Personalized Medicine Comprehensive Cancer Center, Department of Gastroenterology and Digestive Oncology, Hôpital Européen Georges-Pompidou, Paris (J.T.); the Department of Biological Chemistry, National and Kapodistrian University of Athens-School of Medicine, Athens (M.V.K.); Gastrointestinal Oncology Department and Translational Medicine Laboratory, Instituto Nacional de Cancerologia, Mexico City (E.R.-G.); the Oncology Department, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea (T.W.K.); the Medicine Department, Royal Marsden Hospital, London (D.C.); and the Department of Oncology, National Taiwan University Hospital, and the Graduate Institute of Oncology, National Taiwan University College of Medicine - both in Taipei (K.H.Y.)
| | - Michalis V Karamouzis
- From Medical Oncology and Therapeutics Research, City of Hope Comprehensive Cancer Center, Duarte (M.G.F.), and Amgen, Thousand Oaks (E.C., J.C., Y.S., Q.T.) - both in California; Oncologia Medica, Università Cattolica del Sacro Cuore, and Oncologia Medica, Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome (L.S.), the Oncology Department, Fondazione Poliambulanza Istituto Ospedaliero, Brescia (F.M.), the Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa (C.C.), and the Medical Oncology Department, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico, Istituto Nazionale dei Tumori, Milan (F.P.) - all in Italy; the Department of Gastrointestinal and Medical Oncology, National Hospital Organization Kyushu Cancer Center, Fukuoka (T.E.), and the Experimental Therapeutics and GI Oncology Department, National Cancer Center Hospital East, Kashiwa (Y.K.) - both in Japan; the Medicine Department of Hematology, Oncology, and Tumor Immunology, Charité-Universitätsmedizin Berlin, Berlin (D.P.M.); the Department of Medical Oncology, Hospital de la Santa Creu i Sant Pau, Barcelona (D.P.L.-B.); Université Paris Cité, Site de Recherche Intégrée sur le Cancer, Cancer Research for Personalized Medicine Comprehensive Cancer Center, Department of Gastroenterology and Digestive Oncology, Hôpital Européen Georges-Pompidou, Paris (J.T.); the Department of Biological Chemistry, National and Kapodistrian University of Athens-School of Medicine, Athens (M.V.K.); Gastrointestinal Oncology Department and Translational Medicine Laboratory, Instituto Nacional de Cancerologia, Mexico City (E.R.-G.); the Oncology Department, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea (T.W.K.); the Medicine Department, Royal Marsden Hospital, London (D.C.); and the Department of Oncology, National Taiwan University Hospital, and the Graduate Institute of Oncology, National Taiwan University College of Medicine - both in Taipei (K.H.Y.)
| | - Erika Ruiz-Garcia
- From Medical Oncology and Therapeutics Research, City of Hope Comprehensive Cancer Center, Duarte (M.G.F.), and Amgen, Thousand Oaks (E.C., J.C., Y.S., Q.T.) - both in California; Oncologia Medica, Università Cattolica del Sacro Cuore, and Oncologia Medica, Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome (L.S.), the Oncology Department, Fondazione Poliambulanza Istituto Ospedaliero, Brescia (F.M.), the Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa (C.C.), and the Medical Oncology Department, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico, Istituto Nazionale dei Tumori, Milan (F.P.) - all in Italy; the Department of Gastrointestinal and Medical Oncology, National Hospital Organization Kyushu Cancer Center, Fukuoka (T.E.), and the Experimental Therapeutics and GI Oncology Department, National Cancer Center Hospital East, Kashiwa (Y.K.) - both in Japan; the Medicine Department of Hematology, Oncology, and Tumor Immunology, Charité-Universitätsmedizin Berlin, Berlin (D.P.M.); the Department of Medical Oncology, Hospital de la Santa Creu i Sant Pau, Barcelona (D.P.L.-B.); Université Paris Cité, Site de Recherche Intégrée sur le Cancer, Cancer Research for Personalized Medicine Comprehensive Cancer Center, Department of Gastroenterology and Digestive Oncology, Hôpital Européen Georges-Pompidou, Paris (J.T.); the Department of Biological Chemistry, National and Kapodistrian University of Athens-School of Medicine, Athens (M.V.K.); Gastrointestinal Oncology Department and Translational Medicine Laboratory, Instituto Nacional de Cancerologia, Mexico City (E.R.-G.); the Oncology Department, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea (T.W.K.); the Medicine Department, Royal Marsden Hospital, London (D.C.); and the Department of Oncology, National Taiwan University Hospital, and the Graduate Institute of Oncology, National Taiwan University College of Medicine - both in Taipei (K.H.Y.)
| | - Tae-Won Kim
- From Medical Oncology and Therapeutics Research, City of Hope Comprehensive Cancer Center, Duarte (M.G.F.), and Amgen, Thousand Oaks (E.C., J.C., Y.S., Q.T.) - both in California; Oncologia Medica, Università Cattolica del Sacro Cuore, and Oncologia Medica, Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome (L.S.), the Oncology Department, Fondazione Poliambulanza Istituto Ospedaliero, Brescia (F.M.), the Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa (C.C.), and the Medical Oncology Department, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico, Istituto Nazionale dei Tumori, Milan (F.P.) - all in Italy; the Department of Gastrointestinal and Medical Oncology, National Hospital Organization Kyushu Cancer Center, Fukuoka (T.E.), and the Experimental Therapeutics and GI Oncology Department, National Cancer Center Hospital East, Kashiwa (Y.K.) - both in Japan; the Medicine Department of Hematology, Oncology, and Tumor Immunology, Charité-Universitätsmedizin Berlin, Berlin (D.P.M.); the Department of Medical Oncology, Hospital de la Santa Creu i Sant Pau, Barcelona (D.P.L.-B.); Université Paris Cité, Site de Recherche Intégrée sur le Cancer, Cancer Research for Personalized Medicine Comprehensive Cancer Center, Department of Gastroenterology and Digestive Oncology, Hôpital Européen Georges-Pompidou, Paris (J.T.); the Department of Biological Chemistry, National and Kapodistrian University of Athens-School of Medicine, Athens (M.V.K.); Gastrointestinal Oncology Department and Translational Medicine Laboratory, Instituto Nacional de Cancerologia, Mexico City (E.R.-G.); the Oncology Department, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea (T.W.K.); the Medicine Department, Royal Marsden Hospital, London (D.C.); and the Department of Oncology, National Taiwan University Hospital, and the Graduate Institute of Oncology, National Taiwan University College of Medicine - both in Taipei (K.H.Y.)
| | - Yasutoshi Kuboki
- From Medical Oncology and Therapeutics Research, City of Hope Comprehensive Cancer Center, Duarte (M.G.F.), and Amgen, Thousand Oaks (E.C., J.C., Y.S., Q.T.) - both in California; Oncologia Medica, Università Cattolica del Sacro Cuore, and Oncologia Medica, Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome (L.S.), the Oncology Department, Fondazione Poliambulanza Istituto Ospedaliero, Brescia (F.M.), the Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa (C.C.), and the Medical Oncology Department, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico, Istituto Nazionale dei Tumori, Milan (F.P.) - all in Italy; the Department of Gastrointestinal and Medical Oncology, National Hospital Organization Kyushu Cancer Center, Fukuoka (T.E.), and the Experimental Therapeutics and GI Oncology Department, National Cancer Center Hospital East, Kashiwa (Y.K.) - both in Japan; the Medicine Department of Hematology, Oncology, and Tumor Immunology, Charité-Universitätsmedizin Berlin, Berlin (D.P.M.); the Department of Medical Oncology, Hospital de la Santa Creu i Sant Pau, Barcelona (D.P.L.-B.); Université Paris Cité, Site de Recherche Intégrée sur le Cancer, Cancer Research for Personalized Medicine Comprehensive Cancer Center, Department of Gastroenterology and Digestive Oncology, Hôpital Européen Georges-Pompidou, Paris (J.T.); the Department of Biological Chemistry, National and Kapodistrian University of Athens-School of Medicine, Athens (M.V.K.); Gastrointestinal Oncology Department and Translational Medicine Laboratory, Instituto Nacional de Cancerologia, Mexico City (E.R.-G.); the Oncology Department, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea (T.W.K.); the Medicine Department, Royal Marsden Hospital, London (D.C.); and the Department of Oncology, National Taiwan University Hospital, and the Graduate Institute of Oncology, National Taiwan University College of Medicine - both in Taipei (K.H.Y.)
| | - Fausto Meriggi
- From Medical Oncology and Therapeutics Research, City of Hope Comprehensive Cancer Center, Duarte (M.G.F.), and Amgen, Thousand Oaks (E.C., J.C., Y.S., Q.T.) - both in California; Oncologia Medica, Università Cattolica del Sacro Cuore, and Oncologia Medica, Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome (L.S.), the Oncology Department, Fondazione Poliambulanza Istituto Ospedaliero, Brescia (F.M.), the Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa (C.C.), and the Medical Oncology Department, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico, Istituto Nazionale dei Tumori, Milan (F.P.) - all in Italy; the Department of Gastrointestinal and Medical Oncology, National Hospital Organization Kyushu Cancer Center, Fukuoka (T.E.), and the Experimental Therapeutics and GI Oncology Department, National Cancer Center Hospital East, Kashiwa (Y.K.) - both in Japan; the Medicine Department of Hematology, Oncology, and Tumor Immunology, Charité-Universitätsmedizin Berlin, Berlin (D.P.M.); the Department of Medical Oncology, Hospital de la Santa Creu i Sant Pau, Barcelona (D.P.L.-B.); Université Paris Cité, Site de Recherche Intégrée sur le Cancer, Cancer Research for Personalized Medicine Comprehensive Cancer Center, Department of Gastroenterology and Digestive Oncology, Hôpital Européen Georges-Pompidou, Paris (J.T.); the Department of Biological Chemistry, National and Kapodistrian University of Athens-School of Medicine, Athens (M.V.K.); Gastrointestinal Oncology Department and Translational Medicine Laboratory, Instituto Nacional de Cancerologia, Mexico City (E.R.-G.); the Oncology Department, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea (T.W.K.); the Medicine Department, Royal Marsden Hospital, London (D.C.); and the Department of Oncology, National Taiwan University Hospital, and the Graduate Institute of Oncology, National Taiwan University College of Medicine - both in Taipei (K.H.Y.)
| | - David Cunningham
- From Medical Oncology and Therapeutics Research, City of Hope Comprehensive Cancer Center, Duarte (M.G.F.), and Amgen, Thousand Oaks (E.C., J.C., Y.S., Q.T.) - both in California; Oncologia Medica, Università Cattolica del Sacro Cuore, and Oncologia Medica, Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome (L.S.), the Oncology Department, Fondazione Poliambulanza Istituto Ospedaliero, Brescia (F.M.), the Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa (C.C.), and the Medical Oncology Department, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico, Istituto Nazionale dei Tumori, Milan (F.P.) - all in Italy; the Department of Gastrointestinal and Medical Oncology, National Hospital Organization Kyushu Cancer Center, Fukuoka (T.E.), and the Experimental Therapeutics and GI Oncology Department, National Cancer Center Hospital East, Kashiwa (Y.K.) - both in Japan; the Medicine Department of Hematology, Oncology, and Tumor Immunology, Charité-Universitätsmedizin Berlin, Berlin (D.P.M.); the Department of Medical Oncology, Hospital de la Santa Creu i Sant Pau, Barcelona (D.P.L.-B.); Université Paris Cité, Site de Recherche Intégrée sur le Cancer, Cancer Research for Personalized Medicine Comprehensive Cancer Center, Department of Gastroenterology and Digestive Oncology, Hôpital Européen Georges-Pompidou, Paris (J.T.); the Department of Biological Chemistry, National and Kapodistrian University of Athens-School of Medicine, Athens (M.V.K.); Gastrointestinal Oncology Department and Translational Medicine Laboratory, Instituto Nacional de Cancerologia, Mexico City (E.R.-G.); the Oncology Department, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea (T.W.K.); the Medicine Department, Royal Marsden Hospital, London (D.C.); and the Department of Oncology, National Taiwan University Hospital, and the Graduate Institute of Oncology, National Taiwan University College of Medicine - both in Taipei (K.H.Y.)
| | - Kun-Huei Yeh
- From Medical Oncology and Therapeutics Research, City of Hope Comprehensive Cancer Center, Duarte (M.G.F.), and Amgen, Thousand Oaks (E.C., J.C., Y.S., Q.T.) - both in California; Oncologia Medica, Università Cattolica del Sacro Cuore, and Oncologia Medica, Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome (L.S.), the Oncology Department, Fondazione Poliambulanza Istituto Ospedaliero, Brescia (F.M.), the Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa (C.C.), and the Medical Oncology Department, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico, Istituto Nazionale dei Tumori, Milan (F.P.) - all in Italy; the Department of Gastrointestinal and Medical Oncology, National Hospital Organization Kyushu Cancer Center, Fukuoka (T.E.), and the Experimental Therapeutics and GI Oncology Department, National Cancer Center Hospital East, Kashiwa (Y.K.) - both in Japan; the Medicine Department of Hematology, Oncology, and Tumor Immunology, Charité-Universitätsmedizin Berlin, Berlin (D.P.M.); the Department of Medical Oncology, Hospital de la Santa Creu i Sant Pau, Barcelona (D.P.L.-B.); Université Paris Cité, Site de Recherche Intégrée sur le Cancer, Cancer Research for Personalized Medicine Comprehensive Cancer Center, Department of Gastroenterology and Digestive Oncology, Hôpital Européen Georges-Pompidou, Paris (J.T.); the Department of Biological Chemistry, National and Kapodistrian University of Athens-School of Medicine, Athens (M.V.K.); Gastrointestinal Oncology Department and Translational Medicine Laboratory, Instituto Nacional de Cancerologia, Mexico City (E.R.-G.); the Oncology Department, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea (T.W.K.); the Medicine Department, Royal Marsden Hospital, London (D.C.); and the Department of Oncology, National Taiwan University Hospital, and the Graduate Institute of Oncology, National Taiwan University College of Medicine - both in Taipei (K.H.Y.)
| | - Emily Chan
- From Medical Oncology and Therapeutics Research, City of Hope Comprehensive Cancer Center, Duarte (M.G.F.), and Amgen, Thousand Oaks (E.C., J.C., Y.S., Q.T.) - both in California; Oncologia Medica, Università Cattolica del Sacro Cuore, and Oncologia Medica, Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome (L.S.), the Oncology Department, Fondazione Poliambulanza Istituto Ospedaliero, Brescia (F.M.), the Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa (C.C.), and the Medical Oncology Department, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico, Istituto Nazionale dei Tumori, Milan (F.P.) - all in Italy; the Department of Gastrointestinal and Medical Oncology, National Hospital Organization Kyushu Cancer Center, Fukuoka (T.E.), and the Experimental Therapeutics and GI Oncology Department, National Cancer Center Hospital East, Kashiwa (Y.K.) - both in Japan; the Medicine Department of Hematology, Oncology, and Tumor Immunology, Charité-Universitätsmedizin Berlin, Berlin (D.P.M.); the Department of Medical Oncology, Hospital de la Santa Creu i Sant Pau, Barcelona (D.P.L.-B.); Université Paris Cité, Site de Recherche Intégrée sur le Cancer, Cancer Research for Personalized Medicine Comprehensive Cancer Center, Department of Gastroenterology and Digestive Oncology, Hôpital Européen Georges-Pompidou, Paris (J.T.); the Department of Biological Chemistry, National and Kapodistrian University of Athens-School of Medicine, Athens (M.V.K.); Gastrointestinal Oncology Department and Translational Medicine Laboratory, Instituto Nacional de Cancerologia, Mexico City (E.R.-G.); the Oncology Department, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea (T.W.K.); the Medicine Department, Royal Marsden Hospital, London (D.C.); and the Department of Oncology, National Taiwan University Hospital, and the Graduate Institute of Oncology, National Taiwan University College of Medicine - both in Taipei (K.H.Y.)
| | - Joseph Chao
- From Medical Oncology and Therapeutics Research, City of Hope Comprehensive Cancer Center, Duarte (M.G.F.), and Amgen, Thousand Oaks (E.C., J.C., Y.S., Q.T.) - both in California; Oncologia Medica, Università Cattolica del Sacro Cuore, and Oncologia Medica, Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome (L.S.), the Oncology Department, Fondazione Poliambulanza Istituto Ospedaliero, Brescia (F.M.), the Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa (C.C.), and the Medical Oncology Department, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico, Istituto Nazionale dei Tumori, Milan (F.P.) - all in Italy; the Department of Gastrointestinal and Medical Oncology, National Hospital Organization Kyushu Cancer Center, Fukuoka (T.E.), and the Experimental Therapeutics and GI Oncology Department, National Cancer Center Hospital East, Kashiwa (Y.K.) - both in Japan; the Medicine Department of Hematology, Oncology, and Tumor Immunology, Charité-Universitätsmedizin Berlin, Berlin (D.P.M.); the Department of Medical Oncology, Hospital de la Santa Creu i Sant Pau, Barcelona (D.P.L.-B.); Université Paris Cité, Site de Recherche Intégrée sur le Cancer, Cancer Research for Personalized Medicine Comprehensive Cancer Center, Department of Gastroenterology and Digestive Oncology, Hôpital Européen Georges-Pompidou, Paris (J.T.); the Department of Biological Chemistry, National and Kapodistrian University of Athens-School of Medicine, Athens (M.V.K.); Gastrointestinal Oncology Department and Translational Medicine Laboratory, Instituto Nacional de Cancerologia, Mexico City (E.R.-G.); the Oncology Department, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea (T.W.K.); the Medicine Department, Royal Marsden Hospital, London (D.C.); and the Department of Oncology, National Taiwan University Hospital, and the Graduate Institute of Oncology, National Taiwan University College of Medicine - both in Taipei (K.H.Y.)
| | - Yaneth Saportas
- From Medical Oncology and Therapeutics Research, City of Hope Comprehensive Cancer Center, Duarte (M.G.F.), and Amgen, Thousand Oaks (E.C., J.C., Y.S., Q.T.) - both in California; Oncologia Medica, Università Cattolica del Sacro Cuore, and Oncologia Medica, Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome (L.S.), the Oncology Department, Fondazione Poliambulanza Istituto Ospedaliero, Brescia (F.M.), the Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa (C.C.), and the Medical Oncology Department, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico, Istituto Nazionale dei Tumori, Milan (F.P.) - all in Italy; the Department of Gastrointestinal and Medical Oncology, National Hospital Organization Kyushu Cancer Center, Fukuoka (T.E.), and the Experimental Therapeutics and GI Oncology Department, National Cancer Center Hospital East, Kashiwa (Y.K.) - both in Japan; the Medicine Department of Hematology, Oncology, and Tumor Immunology, Charité-Universitätsmedizin Berlin, Berlin (D.P.M.); the Department of Medical Oncology, Hospital de la Santa Creu i Sant Pau, Barcelona (D.P.L.-B.); Université Paris Cité, Site de Recherche Intégrée sur le Cancer, Cancer Research for Personalized Medicine Comprehensive Cancer Center, Department of Gastroenterology and Digestive Oncology, Hôpital Européen Georges-Pompidou, Paris (J.T.); the Department of Biological Chemistry, National and Kapodistrian University of Athens-School of Medicine, Athens (M.V.K.); Gastrointestinal Oncology Department and Translational Medicine Laboratory, Instituto Nacional de Cancerologia, Mexico City (E.R.-G.); the Oncology Department, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea (T.W.K.); the Medicine Department, Royal Marsden Hospital, London (D.C.); and the Department of Oncology, National Taiwan University Hospital, and the Graduate Institute of Oncology, National Taiwan University College of Medicine - both in Taipei (K.H.Y.)
| | - Qui Tran
- From Medical Oncology and Therapeutics Research, City of Hope Comprehensive Cancer Center, Duarte (M.G.F.), and Amgen, Thousand Oaks (E.C., J.C., Y.S., Q.T.) - both in California; Oncologia Medica, Università Cattolica del Sacro Cuore, and Oncologia Medica, Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome (L.S.), the Oncology Department, Fondazione Poliambulanza Istituto Ospedaliero, Brescia (F.M.), the Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa (C.C.), and the Medical Oncology Department, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico, Istituto Nazionale dei Tumori, Milan (F.P.) - all in Italy; the Department of Gastrointestinal and Medical Oncology, National Hospital Organization Kyushu Cancer Center, Fukuoka (T.E.), and the Experimental Therapeutics and GI Oncology Department, National Cancer Center Hospital East, Kashiwa (Y.K.) - both in Japan; the Medicine Department of Hematology, Oncology, and Tumor Immunology, Charité-Universitätsmedizin Berlin, Berlin (D.P.M.); the Department of Medical Oncology, Hospital de la Santa Creu i Sant Pau, Barcelona (D.P.L.-B.); Université Paris Cité, Site de Recherche Intégrée sur le Cancer, Cancer Research for Personalized Medicine Comprehensive Cancer Center, Department of Gastroenterology and Digestive Oncology, Hôpital Européen Georges-Pompidou, Paris (J.T.); the Department of Biological Chemistry, National and Kapodistrian University of Athens-School of Medicine, Athens (M.V.K.); Gastrointestinal Oncology Department and Translational Medicine Laboratory, Instituto Nacional de Cancerologia, Mexico City (E.R.-G.); the Oncology Department, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea (T.W.K.); the Medicine Department, Royal Marsden Hospital, London (D.C.); and the Department of Oncology, National Taiwan University Hospital, and the Graduate Institute of Oncology, National Taiwan University College of Medicine - both in Taipei (K.H.Y.)
| | - Chiara Cremolini
- From Medical Oncology and Therapeutics Research, City of Hope Comprehensive Cancer Center, Duarte (M.G.F.), and Amgen, Thousand Oaks (E.C., J.C., Y.S., Q.T.) - both in California; Oncologia Medica, Università Cattolica del Sacro Cuore, and Oncologia Medica, Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome (L.S.), the Oncology Department, Fondazione Poliambulanza Istituto Ospedaliero, Brescia (F.M.), the Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa (C.C.), and the Medical Oncology Department, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico, Istituto Nazionale dei Tumori, Milan (F.P.) - all in Italy; the Department of Gastrointestinal and Medical Oncology, National Hospital Organization Kyushu Cancer Center, Fukuoka (T.E.), and the Experimental Therapeutics and GI Oncology Department, National Cancer Center Hospital East, Kashiwa (Y.K.) - both in Japan; the Medicine Department of Hematology, Oncology, and Tumor Immunology, Charité-Universitätsmedizin Berlin, Berlin (D.P.M.); the Department of Medical Oncology, Hospital de la Santa Creu i Sant Pau, Barcelona (D.P.L.-B.); Université Paris Cité, Site de Recherche Intégrée sur le Cancer, Cancer Research for Personalized Medicine Comprehensive Cancer Center, Department of Gastroenterology and Digestive Oncology, Hôpital Européen Georges-Pompidou, Paris (J.T.); the Department of Biological Chemistry, National and Kapodistrian University of Athens-School of Medicine, Athens (M.V.K.); Gastrointestinal Oncology Department and Translational Medicine Laboratory, Instituto Nacional de Cancerologia, Mexico City (E.R.-G.); the Oncology Department, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea (T.W.K.); the Medicine Department, Royal Marsden Hospital, London (D.C.); and the Department of Oncology, National Taiwan University Hospital, and the Graduate Institute of Oncology, National Taiwan University College of Medicine - both in Taipei (K.H.Y.)
| | - Filippo Pietrantonio
- From Medical Oncology and Therapeutics Research, City of Hope Comprehensive Cancer Center, Duarte (M.G.F.), and Amgen, Thousand Oaks (E.C., J.C., Y.S., Q.T.) - both in California; Oncologia Medica, Università Cattolica del Sacro Cuore, and Oncologia Medica, Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome (L.S.), the Oncology Department, Fondazione Poliambulanza Istituto Ospedaliero, Brescia (F.M.), the Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa (C.C.), and the Medical Oncology Department, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico, Istituto Nazionale dei Tumori, Milan (F.P.) - all in Italy; the Department of Gastrointestinal and Medical Oncology, National Hospital Organization Kyushu Cancer Center, Fukuoka (T.E.), and the Experimental Therapeutics and GI Oncology Department, National Cancer Center Hospital East, Kashiwa (Y.K.) - both in Japan; the Medicine Department of Hematology, Oncology, and Tumor Immunology, Charité-Universitätsmedizin Berlin, Berlin (D.P.M.); the Department of Medical Oncology, Hospital de la Santa Creu i Sant Pau, Barcelona (D.P.L.-B.); Université Paris Cité, Site de Recherche Intégrée sur le Cancer, Cancer Research for Personalized Medicine Comprehensive Cancer Center, Department of Gastroenterology and Digestive Oncology, Hôpital Européen Georges-Pompidou, Paris (J.T.); the Department of Biological Chemistry, National and Kapodistrian University of Athens-School of Medicine, Athens (M.V.K.); Gastrointestinal Oncology Department and Translational Medicine Laboratory, Instituto Nacional de Cancerologia, Mexico City (E.R.-G.); the Oncology Department, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea (T.W.K.); the Medicine Department, Royal Marsden Hospital, London (D.C.); and the Department of Oncology, National Taiwan University Hospital, and the Graduate Institute of Oncology, National Taiwan University College of Medicine - both in Taipei (K.H.Y.)
| |
Collapse
|
225
|
Tachibana T, Oyama TG, Yoshii Y, Hihara F, Igarashi C, Shinada M, Matsumoto H, Higashi T, Kishimoto T, Taguchi M. An In Vivo Dual-Observation Method to Monitor Tumor Mass and Tumor-Surface Blood Vessels for Developing Anti-Angiogenesis Agents against Submillimeter Tumors. Int J Mol Sci 2023; 24:17234. [PMID: 38139063 PMCID: PMC10743531 DOI: 10.3390/ijms242417234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 11/20/2023] [Accepted: 12/06/2023] [Indexed: 12/24/2023] Open
Abstract
Managing metastasis at the early stage and detecting and treating submillimeter tumors at early metastasis are crucial for improving cancer prognosis. Angiogenesis is a critical target for developing drugs to detect and inhibit submillimeter tumor growth; however, drug development remains challenging because there are no suitable models for observing the submillimeter tumor mass and the surrounding blood vessels in vivo. We have established a xenograft subcutaneous submillimeter tumor mouse model with HT-29-RFP by transplanting a single spheroid grown on radiation-crosslinked gelatin hydrogel microwells. Here, we developed an in vivo dual-observation method to observe the submillimeter tumor mass and tumor-surface blood vessels using this model. RFP was detected to observe the tumor mass, and a fluorescent angiography agent FITC-dextran was administered to observe blood vessels via stereoscopic fluorescence microscopy. The anti-angiogenesis agent regorafenib was used to confirm the usefulness of this method. This method effectively detected the submillimeter tumor mass and tumor-surface blood vessels in vivo. Regorafenib treatment revealed tumor growth inhibition and angiogenesis downregulation with reduced vascular extremities, segments, and meshes. Further, we confirmed that tumor-surface blood vessel areas monitored using in vivo dual-observation correlated with intratumoral blood vessel areas observed via fluorescence microscopy with frozen sections. In conclusion, this method would be useful in developing anti-angiogenesis agents against submillimeter tumors.
Collapse
Affiliation(s)
- Tomoko Tachibana
- Department of Molecular Imaging and Theranostics, National Institutes for Quantum Science and Technology (QST), Chiba 263-8555, Japan; (F.H.); (C.I.); (M.S.); (H.M.); (T.H.)
- Faculty of Science, Toho University, Chiba 274-8510, Japan;
| | - Tomoko Gowa Oyama
- Foundational Quantum Technology Research Directorate, National Institutes for Quantum Science and Technology (QST), Gunma 370-1292, Japan; (T.G.O.); (M.T.)
| | - Yukie Yoshii
- Department of Molecular Imaging and Theranostics, National Institutes for Quantum Science and Technology (QST), Chiba 263-8555, Japan; (F.H.); (C.I.); (M.S.); (H.M.); (T.H.)
- Visible Cancer Drug Research Unit, National Institutes for Quantum Science and Technology (QST), Chiba 263-8555, Japan
| | - Fukiko Hihara
- Department of Molecular Imaging and Theranostics, National Institutes for Quantum Science and Technology (QST), Chiba 263-8555, Japan; (F.H.); (C.I.); (M.S.); (H.M.); (T.H.)
| | - Chika Igarashi
- Department of Molecular Imaging and Theranostics, National Institutes for Quantum Science and Technology (QST), Chiba 263-8555, Japan; (F.H.); (C.I.); (M.S.); (H.M.); (T.H.)
| | - Mitsuhiro Shinada
- Department of Molecular Imaging and Theranostics, National Institutes for Quantum Science and Technology (QST), Chiba 263-8555, Japan; (F.H.); (C.I.); (M.S.); (H.M.); (T.H.)
- Faculty of Science, Toho University, Chiba 274-8510, Japan;
| | - Hiroki Matsumoto
- Department of Molecular Imaging and Theranostics, National Institutes for Quantum Science and Technology (QST), Chiba 263-8555, Japan; (F.H.); (C.I.); (M.S.); (H.M.); (T.H.)
| | - Tatsuya Higashi
- Department of Molecular Imaging and Theranostics, National Institutes for Quantum Science and Technology (QST), Chiba 263-8555, Japan; (F.H.); (C.I.); (M.S.); (H.M.); (T.H.)
| | | | - Mitsumasa Taguchi
- Foundational Quantum Technology Research Directorate, National Institutes for Quantum Science and Technology (QST), Gunma 370-1292, Japan; (T.G.O.); (M.T.)
| |
Collapse
|
226
|
Dolan M, St. John N, Zaidi F, Doyle F, Fasullo M. High-throughput screening of the Saccharomyces cerevisiae genome for 2-amino-3-methylimidazo [4,5-f] quinoline resistance identifies colon cancer-associated genes. G3 (BETHESDA, MD.) 2023; 13:jkad219. [PMID: 37738679 PMCID: PMC11025384 DOI: 10.1093/g3journal/jkad219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 10/25/2022] [Accepted: 09/15/2023] [Indexed: 09/24/2023]
Abstract
Heterocyclic aromatic amines (HAAs) are potent carcinogenic agents found in charred meats and cigarette smoke. However, few eukaryotic resistance genes have been identified. We used Saccharomyces cerevisiae (budding yeast) to identify genes that confer resistance to 2-amino-3-methylimidazo[4,5-f] quinoline (IQ). CYP1A2 and NAT2 activate IQ to become a mutagenic nitrenium compound. Deletion libraries expressing human CYP1A2 and NAT2 or no human genes were exposed to either 400 or 800 µM IQ for 5 or 10 generations. DNA barcodes were sequenced using the Illumina HiSeq 2500 platform and statistical significance was determined for exactly matched barcodes. We identified 424 ORFs, including 337 genes of known function, in duplicate screens of the "humanized" collection for IQ resistance; resistance was further validated for a select group of 51 genes by growth curves, competitive growth, or trypan blue assays. Screens of the library not expressing human genes identified 143 ORFs conferring resistance to IQ per se. Ribosomal protein and protein modification genes were identified as IQ resistance genes in both the original and "humanized" libraries, while nitrogen metabolism, DNA repair, and growth control genes were also prominent in the "humanized" library. Protein complexes identified included the casein kinase 2 (CK2) and histone chaperone (HIR) complex. Among DNA Repair and checkpoint genes, we identified those that function in postreplication repair (RAD18, UBC13, REV7), base excision repair (NTG1), and checkpoint signaling (CHK1, PSY2). These studies underscore the role of ribosomal protein genes in conferring IQ resistance, and illuminate DNA repair pathways for conferring resistance to activated IQ.
Collapse
Affiliation(s)
- Michael Dolan
- College of Nanotechnology, Science, and Engineering, State University of NewYork at Albany, Albany, NY 12203, USA
| | - Nick St. John
- College of Nanotechnology, Science, and Engineering, State University of NewYork at Albany, Albany, NY 12203, USA
| | - Faizan Zaidi
- College of Nanotechnology, Science, and Engineering, State University of NewYork at Albany, Albany, NY 12203, USA
| | - Francis Doyle
- College of Nanotechnology, Science, and Engineering, State University of NewYork at Albany, Albany, NY 12203, USA
| | - Michael Fasullo
- College of Nanotechnology, Science, and Engineering, State University of NewYork at Albany, Albany, NY 12203, USA
| |
Collapse
|
227
|
Tan S, Zhang S, Zhou N, Cai X, Yi C, Gou H. Efficacy and safety of fruquintinib dose-escalation strategy for elderly patients with refractory metastatic colorectal cancer: A single-arm, multicenter, phase II study. Cancer Med 2023; 12:22038-22046. [PMID: 38063405 PMCID: PMC10757135 DOI: 10.1002/cam4.6786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 08/24/2023] [Accepted: 09/30/2023] [Indexed: 12/31/2023] Open
Abstract
BACKGROUND Fruquintinib has demonstrated significant improvement in overall survival (OS) among previously treated metastatic colorectal cancer (mCRC) patients. However, the utilization of fruquintinib has been constrained by various toxicities, such as hand-foot skin reaction (HFSR) and hypertension, particularly in elderly patients with reduced tolerance to the standard dosage. This study aims to investigate the efficacy and safety of fruquintinib dose-escalation strategy for elderly refractory mCRC patients. PATIENTS AND METHODS This open-label, single-arm, phase II trial included patients aged 65 years or over with mCRC who had progressed after two or more lines of chemotherapy. Fruquintinib was administered for 21 consecutive days of a 28-day treatment cycle. The starting dose of fruquintinib was 3 mg/day and escalated to 4 mg/day in Week 2 and 5 mg/day in Week 3 if no significant drug-related toxicity was observed. The highest tolerated dose from Cycle 1 would be administered in Cycle 2 and all subsequent cycles. Before commencing treatment, all enrolled patients underwent a G8 questionnaire and comprehensive geriatric assessments. The primary endpoint of the study was progression-free survival (PFS). RESULTS A total of 29 patients were enrolled and all started fruquintinib at 3 mg/day. Fifteen patients (51.7%) were subsequently escalated to 4 mg/day and 4 (13.8%) to 5 mg/day. Only four (13.8%) patients discontinued treatment due to adverse events (AEs). The median PFS was 3.8 months (95% CI, 2.7-4.9), and the median OS was 7.6 months (95% CI, 6.5-8.7). Treatment-related adverse events (TRAEs) were observed in all 29 patients (100%). The most frequently occurring (>10%) TRAEs greater than Grade 3 were HFSR (20.7%), hypertension (20.7%), and diarrhea (10.3%). CONCLUSION Our study indicated that a dose of 4 mg/day was well tolerated by most elderly patients, suggesting that fruquintinib dose-escalation strategy during the first cycle could serve as a viable alternative to the standard 5 mg/day dosing.
Collapse
Affiliation(s)
- Sirui Tan
- Department of Medical Oncology, Cancer Center, West China HospitalSichuan UniversitySichuanChina
| | - Shunyu Zhang
- Gastric Cancer CenterWest China Hospital, Sichuan UniversitySichuanChina
| | - Nan Zhou
- Gastric Cancer CenterWest China Hospital, Sichuan UniversitySichuanChina
| | - Xiaohong Cai
- Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of MedicineUniversity of Electronic Science and Technology of ChinaChengduChina
| | - Cheng Yi
- Department of Medical Oncology, Cancer Center, West China HospitalSichuan UniversitySichuanChina
| | - Hongfeng Gou
- Department of Medical Oncology, Cancer Center, West China HospitalSichuan UniversitySichuanChina
- Gastric Cancer CenterWest China Hospital, Sichuan UniversitySichuanChina
| |
Collapse
|
228
|
McLoughlin EC, Twamley B, O'Brien JE, Hannon Barroeta P, Zisterer DM, Meegan MJ, O'Boyle NM. Synthesis by diastereomeric resolution, biochemical evaluation and molecular modelling of chiral 3-hydroxyl b-lactam microtubule-targeting agents for the treatment of triple negative breast and chemoresistant colorectal cancers. Bioorg Chem 2023; 141:106877. [PMID: 37804699 DOI: 10.1016/j.bioorg.2023.106877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 09/08/2023] [Accepted: 09/19/2023] [Indexed: 10/09/2023]
Abstract
The synthesis and biochemical activity of a series of chiral trans 3-hydroxyl β-lactams targeting tubulin is described. Synthesis of the series of enantiopure β-lactams was achieved using chiral derivatising reagent N-Boc-l-proline. The absolute configuration was determined as 3S,4S for (+) enantiomer 4EN1 and 3R,4R for (-) enantiomer 4EN2. Antiproliferative studies identified chiral 3S,4S b-lactams with subnanomolar IC50 values across a range of cancer cell lines, improving potency with respect to the corresponding racemates. Fluoro-substituted (+)-(3S,4S)-4-(3-fluoro-4-methoxyphenyl)-3-hydroxy-1-(3,4,5-trimethoxyphenyl)azetidin-2-one (27EN1) was determined as the lead eutomer with dual antiproliferative activity in triple negative breast cancer cells (TNBC), and combretastatin A-4 resistant HT-29 colorectal cancer cells. IC50 values were in the range of 0.26-0.7 nM across four cell lines. Tubulin polymerisation assays, confocal microscopy and molecular modelling studies indicated that 3S,4S eutomers are microtubule destabilisers, while 3R,4R distomers have lower potency as microtubule destabilisers. 27EN1 demonstrated anti-mitotic and pro-apoptotic activity in MDA-MB-231 and HT-29 cells in addition to selective toxicity toward MCF-7 breast cancer versus non-tumorigenic MCF-10-2A cells. The related 3S,4S β-lactam eutomer 4EN1 downregulated expression of key cell survival anti-apoptotic proteins Bcl-2 and Mcl-1 in MDA-MB-231 cells while 27EN1 downregulated Mcl-1 in HT-29 cells. Chiral β-lactam 27EN1 will be further developed for treatment of TNBC and CA-4 resistant colorectal cancers.
Collapse
Affiliation(s)
- Eavan C McLoughlin
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, Panoz Institute and Trinity Biomedical Sciences Institute, 152-160 Pearse Street, Dublin 2, Ireland.
| | - Brendan Twamley
- School of Chemistry, Trinity College Dublin, Dublin 2, Ireland
| | - John E O'Brien
- School of Chemistry, Trinity College Dublin, Dublin 2, Ireland
| | - Patricia Hannon Barroeta
- School of Biochemistry and Immunology, Trinity College Dublin, Trinity Biomedical Sciences Institute, 152-160 Pearse Street, Dublin 2, Ireland
| | - Daniela M Zisterer
- School of Biochemistry and Immunology, Trinity College Dublin, Trinity Biomedical Sciences Institute, 152-160 Pearse Street, Dublin 2, Ireland
| | - Mary J Meegan
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, Panoz Institute and Trinity Biomedical Sciences Institute, 152-160 Pearse Street, Dublin 2, Ireland
| | - Niamh M O'Boyle
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, Panoz Institute and Trinity Biomedical Sciences Institute, 152-160 Pearse Street, Dublin 2, Ireland
| |
Collapse
|
229
|
Çaparlar MA, Durhan A, Süleymanov M, Binarbaşı C, Koşmaz K. Prognostic Effect of Preoperative Inflammatory Markers on Morbidity and Overall Survival in Pancreatic Adenocarsinoma. Niger J Clin Pract 2023; 26:1902-1909. [PMID: 38158359 DOI: 10.4103/njcp.njcp_426_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Accepted: 08/14/2023] [Indexed: 01/03/2024]
Abstract
BACKGROUND/AIM His study aimed to evaluate the availability of common diagnostic tests and biochemical markers in predicting poor prognosis in patients with pancreatic adenocarcinoma (PAC). The primary outcome measure was to identify predictive prognostic factors. The secondary outcome measure was to compare predictive measures in patients who survived or did not survive in the follow-up period. MATERIALS AND METHODS Medical data of 51 patients were obtained who underwent resection surgery for PAC between January 2016 and May 2022. There were two groups according to the mortality in the follow-up period group general mortality positive (GMP; n = 29) and group general mortality negative (GMN; n = 22). Stage IIb was the most common stage in subgroups. RESULTS Preoperative diagnostic tests revealed that aspartate aminotransferase (AST) level, De Ritis ratio (DRR), carcinoembryonic antigen (CEA), cancer antigen 19-9 (CA 19-9), immature granulocyte (IG) count, and IG ratio (IG%) are higher, and hemoglobin (Hgb) levels are lower in the GMP group (P < 0.05). In univariate analysis, seven variables, including AST ≥20.5 (P = 0.001), DRR ≥1.05 (P ≤ 0.001), CEA ≥3.32 (P = 0.02), IG count ≥0.06 (P < 0.01), Hgb ≤11.75 (P = 0.01), poor differentiation (P < 0.001) and existence of life-threatening complication (P < 0.01) were identified. In multivariate analysis, only DRR (≥1.05;100% specificity and 72% sensitivity) and poor differentiation (P = 0.019) were found to be independent prognostic factors for overall survival. The median overall survival of patients with the DRR ≥1.05 and poor tumor differentiation was lower, and the mortality rate was higher than the patients with DRR and without poor tumor differentiation (10.65 ± 3.11 months vs. 61.86 ± 5.36 months and 100% vs. 26.7%, P < 0.001). CONCLUSION Pretreatment high DRR, high IG counts and IG%, and poor differentiation of the tumor might be used as independent predictors of poor prognosis and mortality in patients with PAC.
Collapse
Affiliation(s)
- M A Çaparlar
- Department of General Surgery, Ankara Education and Research Hospital, Ankara, Turkey
| | | | | | | | | |
Collapse
|
230
|
Boshell D, Bester L. Radioembolisation of liver metastases. J Med Imaging Radiat Oncol 2023; 67:842-852. [PMID: 37343147 DOI: 10.1111/1754-9485.13549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Accepted: 06/05/2023] [Indexed: 06/23/2023]
Abstract
This review aims to present contemporary data for SIRT in the treatment of secondary hepatic malignancies including colorectal, neuroendocrine, breast and uveal melanoma.
Collapse
Affiliation(s)
- David Boshell
- Department of Radiology, St Vincent's Hospital, Sydney, New South Wales, Australia
| | - Lourens Bester
- Department of Radiology, University of Notre Dame, Sydney, New South Wales, Australia
| |
Collapse
|
231
|
Jiang DM, Parshad S, Zhan L, Sim HW, Siu LL, Liu G, Shapiro JD, Price TJ, Jonker DJ, Karapetis CS, Strickland AH, Zhang W, Jeffery M, Tu D, Ng S, Sabesan S, Shannon J, Townsend A, O'Callaghan CJ, Chen EX. Plasma Cetuximab Concentrations Correlate With Survival in Patients With Advanced KRAS Wild Type Colorectal Cancer. Clin Colorectal Cancer 2023; 22:457-463. [PMID: 37704538 DOI: 10.1016/j.clcc.2023.08.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 08/18/2023] [Accepted: 08/21/2023] [Indexed: 09/15/2023]
Abstract
BACKGROUND Cetuximab is a standard of care therapy for patients with RAS wild-type (WT) advanced colorectal cancer. Limited data suggest a wide variation in cetuximab plasma concentrations after standard dosing regimens. We correlated cetuximab plasma concentrations with survival and toxicity. METHODS The CO. 20 study randomized patients with RAS WT advanced colorectal cancer in a 1:1 ratio to cetuximab 400 mg/m2 intravenously followed by weekly maintenance of 250 mg/m2, plus brivanib 800 mg orally daily or placebo. Blood samples obtained at week 5 precetuximab treatment were analyzed by ELISA. Patients were grouped into tertiles based on plasma cetuximab concentrations. Cetuximab concentration tertiles were correlated with survival outcomes and toxicity. Patient demographic and biochemical parameters were evaluated as co-variables. RESULTS Week 5 plasma cetuximab concentrations were available for 591 patients (78.8%). The median overall survival (OS) was 11.4 months and 7.8 months for patients in the highest (T3) and lowest tertiles (T1) respectively. On multivariable analysis, plasma cetuximab concentration was associated with OS (HR 0.66, 95% confidence interval [CI]: 0.53-0.83, P < .001, T3 vs. T1), and a trend towards progression-free survival (HR 0.82, 95% CI: 0.66-1.02, P = .07, T3 vs. T1). There was no association between cetuximab concentration and skin toxicity or diarrhea. CONCLUSION The standard cetuximab dosing regimen may not be optimal for all patients. Further pharmacokinetic studies are needed to optimize cetuximab dosing given the potential improvement in OS.
Collapse
Affiliation(s)
- Di Maria Jiang
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto ON
| | - Shruti Parshad
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto ON
| | - Luna Zhan
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto ON
| | - Hao-Wen Sim
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, and NHMRC Clinical Trials Centre, University of Sydney, Sydney, NSW, Ausutralia
| | - Lillian L Siu
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto ON
| | - Geoffrey Liu
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto ON
| | - Jeremy D Shapiro
- Department of Medical Oncology, Cabrini Hospital, Cabrini Monash University, Melbourne, Australia
| | - Timothy J Price
- Department of Hematology and Oncology, Queen Elizabeth Hospital, CALHN, Adelaide, South Australia
| | - Derek J Jonker
- Ottawa Hospital Research Institute, University of Ottawa, Ottawa ON
| | | | | | - Wenjiang Zhang
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto ON
| | - Mark Jeffery
- Canterbury Regional Cancer and Hematology Service Centre, Christchurch Hospital, Christchurch, New Zealand
| | - Dongsheng Tu
- Canadian Cancer Trials Group, Queen's University, Kingston, ON
| | - Siobhan Ng
- Sir Charles Gairdner Hospital, Nedlands, Australia
| | | | | | - Amanda Townsend
- Department of Hematology and Oncology, Queen Elizabeth Hospital, CALHN, Adelaide, South Australia
| | | | - Eric X Chen
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto ON.
| |
Collapse
|
232
|
Ruff SM, Brown ZJ, Pawlik TM. A review of targeted therapy and immune checkpoint inhibitors for metastatic colorectal cancer. Surg Oncol 2023; 51:101993. [PMID: 37742544 DOI: 10.1016/j.suronc.2023.101993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 08/29/2023] [Accepted: 09/17/2023] [Indexed: 09/26/2023]
Abstract
Surgical resection is the cornerstone of treatment for metastatic colorectal cancer (CRC) and offers the best chance at long-term survival. Unfortunately, most patients do not present with resectable metastatic disease and, among patients who do undergo curative-intent resection, many will develop recurrence. In turn, patients require a multi-disciplinary treatment approach with a combination of chemotherapy, surgery, radiation, and/or liver directed therapies that is guided by patient disease burden and clinical status. The development of targeted therapies has led to varying success in other cancers and has emerged as a treatment option for patients with metastatic CRC. While cytotoxic chemotherapy aims to kill cells as they replicate, targeted therapies are directed at biologic features of cancers, like angiogenesis or immune checkpoints. Targeted therapy can facilitate a more treatment tailored approach to the unique genomic alterations of the tumor and hopefully deliver more personalized therapy. We herein provide a systematic review of approved targeted therapies and immune checkpoint inhibitors for metastatic CRC and provide an overview of the current literature.
Collapse
Affiliation(s)
- Samantha M Ruff
- Department of Surgery, Division of Surgical Oncology, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, Columbus, OH, 43210, USA
| | - Zachary J Brown
- Department of Surgery, Division of Surgical Oncology, New York University Long Island, Mineola, NY, USA
| | - Timothy M Pawlik
- Department of Surgery, Division of Surgical Oncology, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, Columbus, OH, 43210, USA.
| |
Collapse
|
233
|
Zhao W, Lei J, Ke S, Chen Y, Xiao J, Tang Z, Wang L, Ren Y, Alnaggar M, Qiu H, Shi W, Yin L, Chen Y. Fecal microbiota transplantation plus tislelizumab and fruquintinib in refractory microsatellite stable metastatic colorectal cancer: an open-label, single-arm, phase II trial (RENMIN-215). EClinicalMedicine 2023; 66:102315. [PMID: 38024475 PMCID: PMC10679864 DOI: 10.1016/j.eclinm.2023.102315] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 10/18/2023] [Accepted: 10/30/2023] [Indexed: 12/01/2023] Open
Abstract
Background Immunotherapy has revolutionized the treatment of cancer. However, microsatellite stable (MSS) metastatic colorectal cancer (mCRC) shows a low response to PD-1 inhibitors. Antiangiogenic therapy can enhance anti-PD-1 efficacy, but it still cannot meet clinical needs. Increasing evidence supported a close relationship between gut microbiome and anti-PD-1 efficacy. This study aimed to explore the efficacy and safety of the combination of fecal microbiota transplantation (FMT) and tislelizumab and fruquintinib in refractory MSS mCRC. Methods In the phase II trial, MSS mCRC patients were administered FMT plus tislelizumab and fruquintinib as a third-line or above treatment. The primary endpoint was progression-free survival (PFS). Secondary endpoints were overall survival (OS), objective response rate (ORR), disease control rate (DCR), duration of response (DoR), clinical benefit rate (CBR), safety and quality of life. Feces and peripheral blood were collected for exploratory biomarker analysis. This study is registered with Chictr.org.cn, identifier ChiCTR2100046768. Findings From May 10, 2021 to January 17, 2022, 20 patients were enrolled. Median follow-up was 13.7 months. Median PFS was 9.6 months (95% CI 4.1-15.1). Median OS was 13.7 months (95% CI 9.3-17.7). Median DoR was 8.1 months (95% CI 1.7-10.6). ORR was 20% (95% CI 5.7-43.7). DCR was 95% (95% CI 75.1-99.9). CBR was 60% (95% CI 36.1-80.9). Nineteen patients (95%) experienced at least one treatment-related adverse event (TRAE). Six patients (30%) had grade 3-4 TRAEs, with the most common being albuminuria (10%), urine occult blood (10%), fecal occult blood (10%), hypertension (5%), hyperglycemia (5%), liver dysfunction (5%), hand-foot skin reaction (5%), and hypothyroidism (5%). No treatment-related deaths occurred. Responders had a high-abundance of Proteobacteria and Lachnospiraceae family and a low-abundance of Actinobacteriota and Bifidobacterium. The treatment did not change the structure of peripheral blood TCR repertoire. However, the expanded TCRs exhibited the characteristics of antigen-driven responses in responders. Interpretation FMT plus tislelizumab and fruquintinib as third-line or above treatment showed improved survival and manageable safety in refractory MSS mCRC, suggesting a valuable new treatment option for this patient population. Funding This study was supported by the National Natural Science Foundation of China (82102954 to Wensi Zhao) and the Special Project of Central Government for Local Science and Technology Development of Hubei Province (ZYYD2020000169 to Yongshun Chen).
Collapse
Affiliation(s)
- Wensi Zhao
- Department of Clinical Oncology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jun Lei
- Department of Clinical Oncology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Shaobo Ke
- Department of Clinical Oncology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yuan Chen
- Department of Clinical Oncology, Qianjiang Central Hospital, Qianjiang, China
| | - Jiping Xiao
- Department of Abdominal Tumor Surgery, Huangshi Central Hospital, Affiliated Hospital of Hubei Polytechnic University, Huangshi, China
| | - Ze Tang
- Department of Abdominal & Pelvic Medical Oncology, Huangshi Central Hospital, Affiliated Hospital of Hubei Polytechnic University, Huangshi, China
| | - Li Wang
- Department of Oncology, Xiaochang First People's Hospital, China
| | - Yiping Ren
- Department of Clinical Oncology, Jingshan Union Hospital of Huazhong University of Science and Technology, Jingshan, China
| | - Mohammed Alnaggar
- Department of Internal Medicine, Clinic Medical College, Hubei University of Science and Technology, Xianning, China
| | - Hu Qiu
- Department of Clinical Oncology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Wei Shi
- Department of Clinical Oncology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Lei Yin
- State Key Laboratory of Virology, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, China
| | - Yongshun Chen
- Department of Clinical Oncology, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
234
|
Honma Y, Ikeda M, Hijioka S, Matsumoto S, Ito T, Aoki T, Furuse J. Optimal first-line treatment strategies of systemic therapy for unresectable gastrointestinal neuroendocrine tumors based on the opinions of Japanese experts. Invest New Drugs 2023; 41:777-786. [PMID: 37856005 DOI: 10.1007/s10637-023-01399-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 10/04/2023] [Indexed: 10/20/2023]
Abstract
There are several options for systemic therapy of gastroenteropancreatic neuroendocrine neoplasms (GEP-NEN), including somatostatin analogues (SSA), molecular-targeted agents, cytotoxic agents, and peptide receptor radionuclide therapy. However, the effectiveness of each agent varies according to the primary site. Although SSA and everolimus are key drugs used for systemic therapy of neuroendocrine tumors arising from the gastrointestinal tract (GI-NET), the optimal strategy for selecting among these modalities remains unexplored. Japanese experts on GI-NET discussed and determined optimal first-line treatment strategies based on the results of previously reported pivotal trials. The consensus was reached that tumor aggressiveness and prognosis can be predicted using hepatic tumor load and Ki-67 labeling index, which are thought to be clinically important factors when selecting systemic therapy for unresectable GI-NET. SSA therapy is considered appropriate for patients with a low hepatic tumor load and low Ki-67 value and everolimus for those with contraindications to SSA therapy. There was also agreement that the treatment strategy should be determined according to whether the origin is in the midgut, considering the biological differences. Based on this strategy, the experts have tentatively created treatment maps and applied them in representative cases of unresectable GI-NET. Japanese experts proposed tentative maps for optimal first-line treatment in patients with unresectable GI-NET. Further investigation is warranted to validate the usefulness of these maps.
Collapse
Affiliation(s)
- Yoshitaka Honma
- Department of Head and Neck, Esophageal Medical Oncology, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan.
| | - Masafumi Ikeda
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, Chiba, 277-8577, Japan
| | - Susumu Hijioka
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Shigemi Matsumoto
- Department of Clinical Oncology, Kyoto University Hospital, 54 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Tetsuhide Ito
- Department of Gastroenterology and Hepatology, International University of Health and Welfare Graduate School of Medicine, Neuroendocrine Tumor Centre, Fukuoka Sanno Hospital, 3-6-45 Momochihama, Sawara-ku, Fukuoka, 814-0001, Japan
| | - Taku Aoki
- Department of Hepato-Biliary-Pancreatic Surgery, Dokkyo Medical University, 800 Kitakobayashi, Mibu, Tochigi, 321-0293, Japan
| | - Junji Furuse
- Kanagawa Cancer Center, 2-3-2 Nakao, Asahi-ku, Yokohama, Kanagawa, 214-8515, Japan
| |
Collapse
|
235
|
Yang L, Atakhanova N, Arellano MTC, Mohamed MY, Hani T, Fahdil AA, Castillo-Acobo RY, Juyal A, Hussein AK, Amin AH, Pecho RDC, Akhavan-Sigari R. Translational research of new developments in targeted therapy of colorectal cancer. Pathol Res Pract 2023; 252:154888. [PMID: 37948996 DOI: 10.1016/j.prp.2023.154888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Revised: 10/08/2023] [Accepted: 10/11/2023] [Indexed: 11/12/2023]
Abstract
A severe global health concern is the rising incidence and mortality rate of colorectal cancer (CRC). Chemotherapy, which is typically used to treat CRC, is known to have limited specificity and can have noticeable side effects. A paradigm shift in cancer treatment has been brought about by the development of targeted therapies, which has led to the appearance of pharmacological agents with improved efficacy and decreased toxicity. Epidermal growth factor receptor (EGFR), vascular endothelial growth factor (VEGF), human epidermal growth factor receptor 2 (HER2), and BRAF are among the molecular targets covered in this review that are used in targeted therapy for CRC. The current discussion also covers advancements in targeted therapeutic approaches, such as antibody-drug conjugates, immune checkpoint inhibitors, and chimeric antigen receptor (CAR) T-cell therapy. A review of the clinical trials and application of these particular therapies in treating CRC is also done. Despite the improvements in targeted therapy for CRC, problems such as drug resistance and patient selection remain to be solved. Despite this, targeted therapies have offered fresh possibilities for identifying and treating CRC, paving the way for the development of personalized medicine and extending the life expectancy and general well-being of CRC patients.
Collapse
Affiliation(s)
- Lei Yang
- Department of Clinical Laboratory, People's Hospital of Chongqing Liangjiang New Area, Chongqing 401121, China
| | - Nigora Atakhanova
- Head of the Department of Oncology, Tashkent Medical Academy, Tashkent 100109, Uzbekistan
| | | | | | - Thamer Hani
- Dentistry Department, Al-Turath University College, Baghdad, Iraq
| | - Ali A Fahdil
- Medical technical college, Al-Farahidi University, Iraq
| | | | - Ashima Juyal
- Uttaranchal Institute of Technology, Uttaranchal University, Dehradun 248007, India
| | | | - Ali H Amin
- Deanship of Scientific Research, Umm Al-Qura University, Makkah, Saudi Arabia
| | | | - Reza Akhavan-Sigari
- Department of Neurosurgery, University Medical Center Tuebingen, Germany; Department of Health Care Management and Clinical Research, Collegium Humanum Warsaw Management University Warsaw, Poland
| |
Collapse
|
236
|
Huang WK, Hsu HC, Yang TS, Lu CW, Pan YR, Wu CE, Chung WH, Hung SI, Yeh CN. Zinc supplementation decreased incidence of grade ≥2 hand-foot skin reaction induced by regorafenib: A phase II randomized clinical trial. Eur J Cancer 2023; 195:113286. [PMID: 37968194 DOI: 10.1016/j.ejca.2023.113286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2023]
Abstract
To the editor: Hand-foot skin reaction (HFSR), characterized by skin abnormalities on palmoplantar surfaces, has an overall incidence of about 35% upon vascular endothelial growth factor receptor-tyrosine kinase inhibitors (VEGFR-TKIs) treatment.1 Zinc, which plays a role in maintaining skin health, may be implicated in the pathogenesis of HFSR.2 Zinc deficiency has been shown to associate with dermatological toxicities of epidermal growth factor receptor (EGFR)-TKI.3, 4 Regorafenib, an oral multi-kinase inhibitor targeting VEGFR 1-3, PDGFR, cKIT, BRAF, and RET1, is approved for the treatment of metastatic colorectal cancer (mCRC) but commonly causes HFSR.5 This phase II randomized trial aimed to investigate whether zinc supplementation can reduce the severity of HFSR induced by regorafenib within the first 8 weeks of treatment (NCT03898102).
Collapse
Affiliation(s)
- Wen-Kuan Huang
- Division of Hematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital, Linkou branch, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Hung-Chih Hsu
- Division of Hematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital, Linkou branch, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Tsai-Sheng Yang
- Division of Hematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital, Linkou branch, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Chun-Wei Lu
- Department of Dermatology, Drug Hypersensitivity Clinical and Research Center, Chang Gung Memorial Hospital, Linkou branch, Taipei, Keelung, Taoyuan, Taiwan; Chang Gung Immunology Consortium, Chang Gung Memorial Hospital, Linkou branch, Taoyuan, Taiwan; Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Kwei-Shan, Taoyuan, Taiwan
| | - Yi-Ru Pan
- Department of General Surgery, GIST team, and Liver Research Center, Chang Gung Memorial Hospital, Linkou branch, Chang Gung University, Taoyuan, Taiwan
| | - Chiao-En Wu
- Division of Hematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital, Linkou branch, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Wen-Hung Chung
- Department of Dermatology, Drug Hypersensitivity Clinical and Research Center, Chang Gung Memorial Hospital, Linkou branch, Taipei, Keelung, Taoyuan, Taiwan; Chang Gung Immunology Consortium, Chang Gung Memorial Hospital, Linkou branch, Taoyuan, Taiwan; Cancer Vaccine and Immune Cell Therapy Core Laboratory, Department of Medical Research, Chang Gung Memorial Hospital, Linkou branch, Taoyuan, Taiwan
| | - Shuen-Iu Hung
- Cancer Vaccine and Immune Cell Therapy Core Laboratory, Department of Medical Research, Chang Gung Memorial Hospital, Linkou branch, Taoyuan, Taiwan; Institute of Pharmacology, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Chun-Nan Yeh
- Department of General Surgery, GIST team, and Liver Research Center, Chang Gung Memorial Hospital, Linkou branch, Chang Gung University, Taoyuan, Taiwan.
| |
Collapse
|
237
|
Nersesian S, Carter EB, Lee SN, Westhaver LP, Boudreau JE. Killer instincts: natural killer cells as multifactorial cancer immunotherapy. Front Immunol 2023; 14:1269614. [PMID: 38090565 PMCID: PMC10715270 DOI: 10.3389/fimmu.2023.1269614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Accepted: 10/30/2023] [Indexed: 12/18/2023] Open
Abstract
Natural killer (NK) cells integrate heterogeneous signals for activation and inhibition using germline-encoded receptors. These receptors are stochastically co-expressed, and their concurrent engagement and signaling can adjust the sensitivity of individual cells to putative targets. Against cancers, which mutate and evolve under therapeutic and immunologic pressure, the diversity for recognition provided by NK cells may be key to comprehensive cancer control. NK cells are already being trialled as adoptive cell therapy and targets for immunotherapeutic agents. However, strategies to leverage their naturally occurring diversity and agility have not yet been developed. In this review, we discuss the receptors and signaling pathways through which signals for activation or inhibition are generated in NK cells, focusing on their roles in cancer and potential as targets for immunotherapies. Finally, we consider the impacts of receptor co-expression and the potential to engage multiple pathways of NK cell reactivity to maximize the scope and strength of antitumor activities.
Collapse
Affiliation(s)
- Sarah Nersesian
- Department of Microbiology and Immunology, Dalhousie University, Halifax, NS, Canada
- Beatrice Hunter Cancer Research Institute, Halifax, NS, Canada
| | - Emily B. Carter
- Department of Microbiology and Immunology, Dalhousie University, Halifax, NS, Canada
- Beatrice Hunter Cancer Research Institute, Halifax, NS, Canada
| | - Stacey N. Lee
- Department of Microbiology and Immunology, Dalhousie University, Halifax, NS, Canada
- Beatrice Hunter Cancer Research Institute, Halifax, NS, Canada
| | | | - Jeanette E. Boudreau
- Department of Microbiology and Immunology, Dalhousie University, Halifax, NS, Canada
- Beatrice Hunter Cancer Research Institute, Halifax, NS, Canada
- Department of Pathology, Dalhousie University, Halifax, NS, Canada
| |
Collapse
|
238
|
Zebrowska K, Banuelos RC, Rizzo EJ, Belk KW, Schneider G, Degeling K. Quantifying the impact of novel metastatic cancer therapies on health inequalities in survival outcomes. Front Pharmacol 2023; 14:1249998. [PMID: 38074129 PMCID: PMC10704132 DOI: 10.3389/fphar.2023.1249998] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 11/10/2023] [Indexed: 03/24/2024] Open
Abstract
Background: Novel therapies in metastatic cancers have contributed to improvements in survival outcomes, yet real-world data suggest that improvements may be mainly driven by those patient groups who already had the highest survival outcomes. This study aimed to develop and apply a framework for quantifying the impact of novel metastatic cancer therapies on health inequalities in survival outcomes based on published aggregate data. Methods: Nine (N = 9) novel therapies for metastatic breast cancer (mBC), metastatic colorectal cancer (mCRC), and metastatic non-small cell lung cancer (mNSCLC) were identified, 3 for each cancer type. Individual patient data (IPD) for overall survival (OS) and progression-free survival (PFS) were replicated from published Kaplan-Meier (KM) curves. For each cancer type, data were pooled for the novel therapies and comparators separately and weighted based on sample size to ensure equal contribution of each therapy in the analyses. Parametric (mixture) distributions were fitted to the weighted data to model and extrapolate survival. The inequality in survival was defined by the absolute difference between groups with the highest and lowest survival for 2 stratifications: one for which survival was stratified into 2 groups and one using 5 groups. Additionally, a linear regression model was fitted to survival estimates for the 5 groups, with the regression coefficient or slope considered as the inequality gradient (IG). The impact of the pooled novel therapies was subsequently defined as the change in survival inequality relative to the pooled comparator therapies. A probabilistic analysis was performed to quantify parameter uncertainty. Results: The analyses found that novel therapies were associated with significant increases in inequalities in survival outcomes relative to their comparators, except in terms of OS for mNSCLC. For mBC, the inequalities in OS increased by 13.9 (95% CI: 1.4; 26.6) months, or 25.0%, if OS was stratified in 5 groups. The IG for mBC increased by 3.2 (0.3; 6.1) months, or 24.7%. For mCRC, inequalities increased by 6.7 (3.0; 10.5) months, or 40.4%, for stratification based on 5 groups; the IG increased by 1.6 (0.7; 2.4) months, or 40.2%. For mNSCLC, inequalities decreased by 14.9 (-84.5; 19.0) months, or 12.2%, for the 5-group stratification; the IG decreased by 2.0 (-16.1; 5.1) months, or 5.5%. Results for the stratification based on 2 groups demonstrated significant increases in OS inequality for all cancer types. In terms of PFS, the increases in survival inequalities were larger in a relative sense compared with OS. For mBC, PFS inequalities increased by 8.7 (5.9; 11.6) months, or 71.7%, for stratification based on 5 groups; the IG increased by 2.0 (1.3; 2.6) months, or 67.6%. For mCRC, PFS inequalities increased by 5.4 (4.2; 6.6) months, or 147.6%, for the same stratification. The IG increased by 1.3 (1.1; 1.6) months, or 172.7%. For mNSCLC, inequalities increased by 18.2 (12.5; 24.4) months, or 93.8%, for the 5-group stratification; the IG increased by 4.0 (2.8; 5.4) months, or 88.1%. Results from the stratification based on 2 groups were similar. Conclusion: Novel therapies for mBC, mCRC, and mNSCLC are generally associated with significant increases in survival inequalities relative to their comparators in randomized controlled trials, though inequalities in OS for mNSCLC decreased nonsignificantly when stratified based on 5 groups. Although further research using real-world IPD is warranted to assess how, for example, social determinants of health affect the impact of therapies on health inequalities among patient groups, the proposed framework can provide important insights in the absence of such data.
Collapse
Affiliation(s)
| | | | | | - Kathy W. Belk
- Healthcare Consultancy Group, New York, NY, United States
| | - Gary Schneider
- Healthcare Consultancy Group, New York, NY, United States
| | - Koen Degeling
- Healthcare Consultancy Group, London, United Kingdom
| |
Collapse
|
239
|
Bitsianis S, Mantzoros I, Anestiadou E, Christidis P, Chatzakis C, Zapsalis K, Symeonidis S, Ntampakis G, Domvri K, Tsakona A, Bekiari C, Ioannidis O, Aggelopoulos S. Effect of Intraperitoneal Chemotherapy with Regorafenib on IL-6 and TNF-α Levels and Peritoneal Cytology: Experimental Study in Rats with Colorectal Peritoneal Carcinomatosis. J Clin Med 2023; 12:7267. [PMID: 38068319 PMCID: PMC10706907 DOI: 10.3390/jcm12237267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 11/16/2023] [Accepted: 11/22/2023] [Indexed: 08/11/2024] Open
Abstract
Cytoreductive surgery (CRS), combined with hyperthermic intraperitoneal chemotherapy, has significantly improved survival outcomes in patients with peritoneal carcinomatosis from colorectal cancer (CRC). Regorafenib is an oral agent administered in patients with refractory metastatic CRC. Our aim was to investigate the outcomes of intraperitoneal administration of regorafenib for intraperitoneal chemotherapy (IPEC) or/and CRS in a rat model of colorectal peritoneal metastases regarding immunology and peritoneal cytology. A total of 24 rats were included. Twenty-eight days after carcinogenesis induction, rats were randomized into following groups: group A: control group; group B: CRS only; group C: IPEC only; and group D: CRS + IPEC. On day 56 after carcinogenesis, euthanasia and laparotomy were performed. Serum levels of interleukin-6 (IL-6) and tumor necrosis factor α (TNF-α) as well as peritoneal cytology were investigated. Groups B and D had statistically significant lower mean levels of IL-6 and TNF-α compared to groups A and C, but there was no significant difference between them. Both B and D groups presented a statistically significant difference regarding the rate of negative peritoneal cytology, when compared to the control group, but not to group C. In conclusion, regorafenib-based IPEC, combined with CRS, may constitute a promising tool against peritoneal carcinomatosis by altering the tumor microenvironment.
Collapse
Affiliation(s)
- Stefanos Bitsianis
- 4th Department of General Surgery, Aristotle University of Thessaloniki, General Hospital of Thessaloniki “G. Papanikolaou”, 54124 Thessaloniki, Greece; (S.B.); (I.M.); (E.A.); (P.C.); (C.C.); (K.Z.); (S.S.); (G.N.); (S.A.)
| | - Ioannis Mantzoros
- 4th Department of General Surgery, Aristotle University of Thessaloniki, General Hospital of Thessaloniki “G. Papanikolaou”, 54124 Thessaloniki, Greece; (S.B.); (I.M.); (E.A.); (P.C.); (C.C.); (K.Z.); (S.S.); (G.N.); (S.A.)
| | - Elissavet Anestiadou
- 4th Department of General Surgery, Aristotle University of Thessaloniki, General Hospital of Thessaloniki “G. Papanikolaou”, 54124 Thessaloniki, Greece; (S.B.); (I.M.); (E.A.); (P.C.); (C.C.); (K.Z.); (S.S.); (G.N.); (S.A.)
| | - Panagiotis Christidis
- 4th Department of General Surgery, Aristotle University of Thessaloniki, General Hospital of Thessaloniki “G. Papanikolaou”, 54124 Thessaloniki, Greece; (S.B.); (I.M.); (E.A.); (P.C.); (C.C.); (K.Z.); (S.S.); (G.N.); (S.A.)
| | - Christos Chatzakis
- 4th Department of General Surgery, Aristotle University of Thessaloniki, General Hospital of Thessaloniki “G. Papanikolaou”, 54124 Thessaloniki, Greece; (S.B.); (I.M.); (E.A.); (P.C.); (C.C.); (K.Z.); (S.S.); (G.N.); (S.A.)
| | - Konstantinos Zapsalis
- 4th Department of General Surgery, Aristotle University of Thessaloniki, General Hospital of Thessaloniki “G. Papanikolaou”, 54124 Thessaloniki, Greece; (S.B.); (I.M.); (E.A.); (P.C.); (C.C.); (K.Z.); (S.S.); (G.N.); (S.A.)
| | - Savvas Symeonidis
- 4th Department of General Surgery, Aristotle University of Thessaloniki, General Hospital of Thessaloniki “G. Papanikolaou”, 54124 Thessaloniki, Greece; (S.B.); (I.M.); (E.A.); (P.C.); (C.C.); (K.Z.); (S.S.); (G.N.); (S.A.)
| | - Georgios Ntampakis
- 4th Department of General Surgery, Aristotle University of Thessaloniki, General Hospital of Thessaloniki “G. Papanikolaou”, 54124 Thessaloniki, Greece; (S.B.); (I.M.); (E.A.); (P.C.); (C.C.); (K.Z.); (S.S.); (G.N.); (S.A.)
| | - Kalliopi Domvri
- Laboratory of Histology-Embryology, Medical School, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece;
- Department of Pathology, General Hospital of Thessaloniki “G. Papanikolaou”, 54124 Thessaloniki, Greece
| | - Anastasia Tsakona
- Pathology Department, Faculty of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece;
| | - Chryssa Bekiari
- Experimental and Research Center, Papageorgiou General Hospital of Thessaloniki, 56403 Thessaloniki, Greece;
- Laboratory of Anatomy and Histology, Veterinary School, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Orestis Ioannidis
- 4th Department of General Surgery, Aristotle University of Thessaloniki, General Hospital of Thessaloniki “G. Papanikolaou”, 54124 Thessaloniki, Greece; (S.B.); (I.M.); (E.A.); (P.C.); (C.C.); (K.Z.); (S.S.); (G.N.); (S.A.)
| | - Stamatios Aggelopoulos
- 4th Department of General Surgery, Aristotle University of Thessaloniki, General Hospital of Thessaloniki “G. Papanikolaou”, 54124 Thessaloniki, Greece; (S.B.); (I.M.); (E.A.); (P.C.); (C.C.); (K.Z.); (S.S.); (G.N.); (S.A.)
| |
Collapse
|
240
|
Mullin G, Sternschuss M, Landman Y, Sulkes A, Brenner B. Mitomycin C and capecitabine: An additional option as an advanced line therapy in patients with metastatic colorectal cancer. World J Gastrointest Oncol 2023; 15:1913-1924. [DOI: 10.4251/wjgo.v15.i11.1913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 08/31/2023] [Accepted: 10/11/2023] [Indexed: 11/15/2023] Open
Abstract
BACKGROUND In recent years survival of patients with metastatic colorectal cancer (mCRC), though still limited, has improved significantly; clearly, when the disease becomes refractory to standard regimens, additional treatment options are needed. Studies have shown that mitomycin C (MMC), an antitumor antibiotic, and capecitabine, a precursor of 5-fluorouracil, may act synergistically in combination. The efficacy of MMC/capecitabine has been demonstrated in the first-line setting, but only a few small studies have tested it in the advanced-line setting, with contradictory results.
AIM To summarize our experience using MMC/capecitabine as an advanced line treatment for mCRC.
METHODS A retrospective study was conducted at a tertiary medical center including all patients with histologically proven mCRC who were treated with MMC/capecitabine after at least two previous lines of standard chemotherapy in 2006-2020. Data on patient demographics and past medical history, laboratory, pathological, and radiological factors, and treatment and survival were collected from the files. Survival analyses were performed using the Kaplan-Meier method. The association of patient and tumor characteristics with treatment effectiveness and toxicity was evaluated with univariate and multivariate proportional hazard Cox regression analyses. P ≤ 0.05 was considered statistically significant.
RESULTS The cohort consisted of 119 patients of median age 64 years (range 37-85). Patients received a median of 2 MMC/capecitabine cycles (range 0.5-9.0). Thirty-four patients (28.6%) experienced grade ≥ 3 toxicity, including 2 (1.7%) with grade 4; there was no drug-related mortality. The objective response rate was 0.8%, and the disease control rate, 24.4%. Median progression-free survival (PFS) was 2.1 mo (range 0.2-20.3), and median overall survival, 4.8 mo (range 0.2-27.5). The 6-month overall survival rate was 44%; 8.7% of patients remained progression-free. Factors associated with longer PFS were lower gamma-glutamyl transferase level (P = 0.030) and primary tumor location in the left colon (P = 0.017). Factors associated with longer overall survival were lower gamma-glutamyl transferase level (P = 0.022), left-colon tumor location (P = 0.044), low-to-moderate histological grade (P = 0.012), Eastern Cooperative Oncology Group performance status 0-1 (P = 0.036), and normal bilirubin level (P = 0.047).
CONCLUSION MMC/capecitabine is an active, available, and relatively safe regimen for use beyond standard lines of therapy in mCRC. Several clinical and laboratory parameters can identify patients more likely to benefit.
Collapse
Affiliation(s)
- Gil Mullin
- Faculty of Medicine, Tel-Aviv University, Tel Aviv 6997801, Israel
- Institute of Oncology, Davidoff Cancer Center, Beilinson Campus, Petah-Tikva 4941492, Israel
| | - Michal Sternschuss
- Institute of Oncology, Davidoff Cancer Center, Beilinson Campus, Petah-Tikva 4941492, Israel
| | - Yosef Landman
- Institute of Oncology, Davidoff Cancer Center, Beilinson Campus, Petah-Tikva 4941492, Israel
| | - Aaron Sulkes
- Faculty of Medicine, Tel-Aviv University, Tel Aviv 6997801, Israel
- Institute of Oncology, Davidoff Cancer Center, Beilinson Campus, Petah-Tikva 4941492, Israel
| | - Baruch Brenner
- Faculty of Medicine, Tel-Aviv University, Tel Aviv 6997801, Israel
- Institute of Oncology, Davidoff Cancer Center, Beilinson Campus, Petah-Tikva 4941492, Israel
| |
Collapse
|
241
|
Wang D, Ge H, Lu Y, Gong X. Incidence trends and survival analysis of appendiceal tumors in the United States: Primarily changes in appendiceal neuroendocrine tumors. PLoS One 2023; 18:e0294153. [PMID: 37956190 PMCID: PMC10642837 DOI: 10.1371/journal.pone.0294153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 10/25/2023] [Indexed: 11/15/2023] Open
Abstract
BACKGROUND Appendiceal tumors are considered to be a relatively rare tumor of the gastrointestinal tract and the prognosis is unclear. This study comprehensively investigated trends in the epidemiology and survival of appendiceal tumors in the United States over the past approximately 20 years. METHODS Patients with pathologically confirmed appendiceal tumors from 2000 to 2017 were selected from the Surveillance, Epidemiology and End Results (SEER) database. Age-adjusted incidence rates were calculated by SEER*Stat 8.4.0. The Kaplan-Meier method was used to analyze survival and prognostic factors were investigated by a multivariate Cox proportional risk model. RESULTS Ultimately, 13,546 patients with appendiceal tumors between 2000 and 2017 were included. The annual incidence of colonic adenocarcinoma and mucinous adenocarcinoma remained relatively stable. Interestingly, the annual incidence of appendiceal neuroendocrine tumors (aNETs) increased significantly, from 0.03 to 0.90 per 100,000 person-years, with the most dramatic increase in the number of patients with localized disease. Patients with aNETs showed a significant improvement in survival between 2009-2017, compared to the period 2000-2008. Moreover, this improvement in survival over time was seen at all stages (localized, regional, distant) of aNETs. However, this improved survival over time was not seen in colonic and mucinous adenocarcinoma. CONCLUSIONS The incidence of appendiceal neoplasms has increased significantly over the past nearly two decades, which is mainly due to the increased incidence and significant migration to earlier stages in aNETs. We must note that despite the increased incidence of aNETs, survival rates have improved at different disease stages.
Collapse
Affiliation(s)
- Dan Wang
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Heming Ge
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Department of General Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Yebin Lu
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Xuejun Gong
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
242
|
Del Rivero J, Perez K, Kennedy EB, Mittra ES, Vijayvergia N, Arshad J, Basu S, Chauhan A, Dasari AN, Bellizzi AM, Gangi A, Grady E, Howe JR, Ivanidze J, Lewis M, Mailman J, Raj N, Soares HP, Soulen MC, White SB, Chan JA, Kunz PL, Singh S, Halfdanarson TR, Strosberg JR, Bergsland EK. Systemic Therapy for Tumor Control in Metastatic Well-Differentiated Gastroenteropancreatic Neuroendocrine Tumors: ASCO Guideline. J Clin Oncol 2023; 41:5049-5067. [PMID: 37774329 DOI: 10.1200/jco.23.01529] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 07/24/2023] [Indexed: 10/01/2023] Open
Abstract
PURPOSE To develop recommendations for systemic therapy for well-differentiated grade 1 (G1) to grade 3 (G3) metastatic gastroenteropancreatic neuroendocrine tumors (GEP-NETs). METHODS ASCO convened an Expert Panel to conduct a systematic review of relevant studies and develop recommendations for clinical practice. RESULTS Eight randomized controlled trials met the inclusion criteria for the systematic review. RECOMMENDATIONS Somatostatin analogs (SSAs) are recommended as first-line systemic therapy for most patients with G1-grade 2 (G2) metastatic well-differentiated GI-NETs. Observation is an option for patients with low-volume or slow-growing disease without symptoms. After progression on SSAs, peptide receptor radionuclide therapy (PRRT) is recommended as systematic therapy for patients with somatostatin receptor (SSTR)-positive tumors. Everolimus is an alternative second-line therapy, particularly in nonfunctioning NETs and patients with SSTR-negative tumors. SSAs are standard first-line therapy for SSTR-positive pancreatic (pan)NETs. Rarely, observation may be appropriate for asymptomatic patients until progression. Second-line systemic options for panNETs include PRRT (for SSTR-positive tumors), cytotoxic chemotherapy, everolimus, or sunitinib. For SSTR-negative tumors, first-line therapy options are chemotherapy, everolimus, or sunitinib. There are insufficient data to recommend particular sequencing of therapies. Patients with G1-G2 high-volume disease, relatively high Ki-67 index, and/or symptoms related to tumor growth may benefit from early cytotoxic chemotherapy. For G3 GEP-NETs, systemic options for G1-G2 may be considered, although cytotoxic chemotherapy is likely the most effective option for patients with tumor-related symptoms, and SSAs are relatively ineffective. Qualifying statements are provided to assist with treatment choice. Multidisciplinary team management is recommended, along with shared decision making with patients, incorporating their values and preferences, potential benefits and harms, and other characteristics and circumstances, such as comorbidities, performance status, geographic location, and access to care.Additional information is available at www.asco.org/gastrointestinal-cancer-guidelines.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Sandip Basu
- Bhabha Atomic Research Centre, Tata Memorial Hospital, Mumbai, India
| | | | | | | | | | | | | | | | | | | | - Nitya Raj
- Memorial Sloan Kettering Cancer Center, New York, NY
| | | | | | | | | | | | - Simron Singh
- Odette Cancer Center, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | | | | | | |
Collapse
|
243
|
Aydin SG, Kavak EE, Topcu A, Bayramgil A, Akgul F, Kahraman S, Aykan MB, Altıntas YE, Helvaci K, Urun Y, Bilici A, Seker M, Nahit Sendur MA, Olmez OF, Acikgoz O, Cicin I. Prognostic factors for regorafenib treatment in patients with refractory metastatic colorectal cancer: A real-life retrospective multi-center study. BIOMOLECULES & BIOMEDICINE 2023; 23:1089-1095. [PMID: 37289436 PMCID: PMC10655877 DOI: 10.17305/bb.2023.9253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Revised: 06/02/2023] [Accepted: 06/02/2023] [Indexed: 06/09/2023]
Abstract
Regorafenib, an oral multikinase inhibitor, has improved survival in metastatic colorectal cancer (mCRC) patients who have progressed on standard therapies. Our study aimed to evaluate prognostic factors influencing regorafenib treatment and assess the optimal dosing regimen in a real-life setting. We retrospectively analysed 263 patients with mCRC from multiple medical oncology clinics in Turkey. Treatment responses and prognostic factors for survival were evaluated using univariate and multivariate analysis. Of the patients, 120 were male, and 143 were female; 28.9% of tumors were located in the rectum. RAS mutations were present in 3.0% of tumors, while BRAF, K-RAS, and N-RAS mutations were found in 3.0%, 29.7%, and 25.9% of tumor tissues, respectively. Dose escalation was preferred in 105 (39.9%) patients. The median treatment duration was 3.0 months, with an objective response rate (ORR) of 4.9%. Grade ≥ 3 treatment-related toxicity occurred in 133 patients, leading to discontinuation, interruption, and modification rates of 50.6%, 43.7%, and 79.0%, respectively. Median progression-free survival (PFS) and overall survival (OS) were 3.0 and 8.1 months, respectively. RAS/RAF mutation (hazard ratio [HR] 1.5, 95% confidence interval [CI] 1.1-2.3; P = 0.01), pretreatment carcinoembryonic antigen (CEA) levels (HR 1.6, 95% CI 1.1-2.3; P = 0.008), and toxicity-related treatment interruption or dose adjustment (HR 1.6, 95% CI 1.1-2.4; P = 0.01) were identified as independent prognostic factors for PFS. Dose escalation had no significant effect on PFS but was associated with improved OS (P < 0.001). Independent prognostic factors for OS were the initial TNM stage (HR 1.3, 95% CI 1.0-1.9; P = 0.04) and dose interruption/adjustment (HR 0.4, 95% CI 0.2-0.9; P = 0.03). Our findings demonstrate the efficacy and safety of regorafenib. Treatment line influences the response, with dose escalation being more favorable than adjustment or interruption, thus impacting survival.
Collapse
Affiliation(s)
- Sabin Goktas Aydin
- Department of Medical Oncology, Medical Faculty, Medipol University, Istanbul, Turkey
| | - Engin Eren Kavak
- Department of Medical Oncology, Medical Faculty, Ankara University, Ankara, Turkey
| | - Atakan Topcu
- Department of Medical Oncology, Medical Faculty, Bezmialem Vakif University, Istanbul, Turkey
| | - Ayberk Bayramgil
- Department of Medical Oncology, Medical Faculty, Medipol University, Istanbul, Turkey
| | - Fahri Akgul
- Department of Medical Oncology, Medical Faculty, Trakya University, Edirne, Turkey
| | - Seda Kahraman
- Department of Medical Oncology, Medical Faculty, Ankara Yildirim Beyazit University, Ankara, Turkey
| | - Musa Baris Aykan
- Department of Medical Oncology, Gulhane Education and Research Hospital, Ankara, Turkey
| | - Yunus Emre Altıntas
- Department of Medical Oncology, Medical Faculty, Koc University, Istanbul, Turkey
| | - Kaan Helvaci
- Department of Medical Oncology, Memorial Ankara Hospital, Ankara, Turkey
| | - Yuksel Urun
- Department of Medical Oncology, Medical Faculty, Ankara University, Ankara, Turkey
| | - Ahmet Bilici
- Department of Medical Oncology, Medical Faculty, Medipol University, Istanbul, Turkey
| | - Mesut Seker
- Department of Medical Oncology, Medical Faculty, Bezmialem Vakif University, Istanbul, Turkey
| | - Mehmet Ali Nahit Sendur
- Department of Medical Oncology, Medical Faculty, Ankara Yildirim Beyazit University, Ankara, Turkey
| | - Omer Fatih Olmez
- Department of Medical Oncology, Medical Faculty, Medipol University, Istanbul, Turkey
| | - Ozgur Acikgoz
- Department of Medical Oncology, Medical Faculty, Medipol University, Istanbul, Turkey
| | - Irfan Cicin
- Department of Medical Oncology, Medical Faculty, Trakya University, Edirne, Turkey
| |
Collapse
|
244
|
Oh DY, Maqueda MA, Quinn DI, O'Dwyer PJ, Chau I, Kim SY, Duran I, Castellano D, Berlin J, Mellado B, Williamson SK, Lee KW, Marti F, Mathew P, Saif MW, Wang D, Chong E, Hilger-Rolfe J, Dean JP, Arkenau HT. Ibrutinib combination therapy for advanced gastrointestinal and genitourinary tumours: results from a phase 1b/2 study. BMC Cancer 2023; 23:1056. [PMID: 37919668 PMCID: PMC10623721 DOI: 10.1186/s12885-023-11539-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 10/18/2023] [Indexed: 11/04/2023] Open
Abstract
BACKGROUND Ibrutinib, a first-in-class inhibitor of Bruton's tyrosine kinase, is approved for the treatment of various B-cell malignancies and chronic graft-versus-host disease. Based on encouraging preclinical data, safety and efficacy of ibrutinib combined with companion drugs for advanced renal cell carcinoma (RCC), gastric/gastroesophageal junctional adenocarcinoma (GC), and colorectal adenocarcinoma (CRC) were evaluated. METHODS Ibrutinib 560 mg or 840 mg once daily was administered with standard doses of everolimus for RCC, docetaxel for GC, and cetuximab for CRC. Endpoints included determination of the recommended phase 2 dose (RP2D) of ibrutinib in phase 1b and efficacy (overall response rate [ORR] for GC and CRC; progression-free survival [PFS] for CRC) in phase 2. RESULTS A total of 39 (RCC), 46 (GC), and 50 (RCC) patients were enrolled and received the RP2D. Safety profiles were consistent with the individual agents used in the study. Confirmed ORRs were 3% (RCC), 21% (GC), and 19% (CRC). Median (90% CI) PFS was 5.6 (3.9-7.5) months in RCC, 4.0 (2.7-4.2) months in GC, and 5.4 (4.1-5.8) months in CRC. CONCLUSIONS Clinically meaningful increases in efficacy were not observed compared to historical controls; however, the data may warrant further evaluation of ibrutinib combinations in other solid tumours. TRIAL REGISTRATION ClinicalTrials.gov, NCT02599324.
Collapse
Affiliation(s)
- Do-Youn Oh
- Seoul National University Hospital, Cancer Research Institute, Seoul National University College of Medicine, Seoul National University Graduate School, Seoul, South Korea.
| | | | - David I Quinn
- University of Southern California Norris Comprehensive Cancer Center, Los Angeles, CA, USA
| | | | - Ian Chau
- The Royal Marsden NHS Foundation Trust-Royal Marsden Hospital, London, UK
| | - Sun Young Kim
- Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Ignacio Duran
- Hospital Universitario Marques de Valdecilla, IDIVAL, Santander, Spain
| | | | | | - Begona Mellado
- Medical Oncology Department, Hospital Clinic i Provincial de Barcelona, IDIBAPS, University of Barcelona, Barcelona, Spain
| | | | - Keun-Wook Lee
- Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea
| | | | | | - Muhammad Wasif Saif
- Tufts Medical Center, Boston, MA, USA
- Orlando Health Cancer Institute, Orlando, FL, USA
| | - Ding Wang
- Henry Ford Hospital, Detroit, MI, USA
| | - Elizabeth Chong
- Pharmacyclics LLC, an AbbVie Company, South San Francisco, CA, USA
| | | | - James P Dean
- Pharmacyclics LLC, an AbbVie Company, South San Francisco, CA, USA
| | - Hendrik-Tobias Arkenau
- Sarah Cannon Research Institute - United Kingdom (SCRI-UK) and University College London, Cancer Institute, London, UK
| |
Collapse
|
245
|
Kobecki J, Gajdzis P, Mazur G, Chabowski M. Prognostic Potential of Nectin Expressions in Colorectal Cancer: An Exploratory Study. Int J Mol Sci 2023; 24:15900. [PMID: 37958883 PMCID: PMC10650805 DOI: 10.3390/ijms242115900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 10/17/2023] [Accepted: 10/31/2023] [Indexed: 11/15/2023] Open
Abstract
Colorectal cancer (CRC) is a pressing global health challenge, with an estimated 1.9 million new cases in 2020. Ranking as the third most diagnosed cancer globally, CRC accounts for nearly 930,000 cancer-related deaths annually. Nectins, immunoglobulin-like adhesion molecules, are pivotal in intercellular adhesion formation and cellular function regulation. Altered nectin expression patterns have been identified in various cancers. However, the intricacies of their role in cancer development and progression remain underexplored. This study aimed to evaluate the expression of specific nectins in CRC tumors, explore their association with clinicopathological factors, and ascertain their potential as prognostic indicators for CRC patients post-resection. We retrospectively analyzed the medical records of 92 CRC patients who underwent surgical treatment between 2013 and 2014. Tumor specimens were re-evaluated to determine nectin expression using immunohistochemistry. The study identified heterogeneous expressions of nectin-2, -3, and -4 in 58%, 62.6%, and 87.9% of specimens, respectively. Elevated nectin-4 expression correlated with worse 5-year and overall survival rates, presenting a negative prognostic value (HR = 4, 95% CI: 2.4-6.8, p < 0.001). Conversely, reduced nectin-3 expression was linked to poorer CRC prognosis (HR = 0.54; 95% CI: 0.31-0.96; p = 0.036). Nectin-4 expression positively correlated with elevated carcinoembryonic antigen (CEA) levels and advanced disease stages. In contrast, nectin-3 expression negatively correlated with CEA levels, tumor size, presence of distant metastases, and disease stage. Notably, tumors in the right colon were statistically more likely to express nectin-2 compared to those in the left. This study underscores the potential prognostic significance of nectins in CRC. The high prevalence of nectin-4-expressing cells offers promising avenues for further evaluation in targeted therapeutic interventions with already available agents such as PADCEV.
Collapse
Affiliation(s)
- Jakub Kobecki
- Department of Surgery, 4th Military Teaching Hospital, 5 Weigla Street, 50-981 Wroclaw, Poland;
- Division of Anaesthesiological and Surgical Nursing, Department of Nursing and Obstetrics, Faculty of Health Science, Wroclaw Medical University, 5 Bartla Street, 51-618 Wroclaw, Poland
| | - Paweł Gajdzis
- Department of Pathomorphology, 4th Military Teaching Hospital, 5 Weigla Street, 50-981 Wroclaw, Poland;
- Department of Clinical Pathology, Wroclaw Medical University, 213 Borowska Street, 50-556 Wroclaw, Poland
| | - Grzegorz Mazur
- Department of Internal Medicine, Occupational Diseases, Hypertension and Clinical Oncology, Wroclaw Medical University, 213 Borowska Street, 50-556 Wroclaw, Poland;
| | - Mariusz Chabowski
- Department of Surgery, 4th Military Teaching Hospital, 5 Weigla Street, 50-981 Wroclaw, Poland;
- Division of Anaesthesiological and Surgical Nursing, Department of Nursing and Obstetrics, Faculty of Health Science, Wroclaw Medical University, 5 Bartla Street, 51-618 Wroclaw, Poland
- Department of Clinical Surgical Sciences, Faculty of Medicine, Wroclaw University of Science and Technology, 50-556 Wroclaw, Poland
| |
Collapse
|
246
|
Kim KH, Jung M, Lee HJ, Lee SJ, Kim M, Ahn MS, Choi MY, Lee NR, Shin SJ. A phase II study on the efficacy of regorafenib in treating patients with c-KIT-mutated metastatic malignant melanoma that progressed after previous treatment (KCSG-UN-14-13). Eur J Cancer 2023; 193:113312. [PMID: 37741071 DOI: 10.1016/j.ejca.2023.113312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 08/14/2023] [Accepted: 08/22/2023] [Indexed: 09/25/2023]
Abstract
BACKGROUND c-KIT mutations are found in approximately 15% of patients with malignant melanoma in the Asian population. Regorafenib, an oral multikinase inhibitor, acts against both wild-type and mutant KIT. OBJECTIVE This multi-institutional, phase II, single-arm study aimed to evaluate the efficacy of regorafenib against metastatic malignant melanoma harbouring c-KIT mutations. METHODS Patients with metastatic melanoma positive for c-KIT mutations, upon progression after at least one line of systemic treatment, were enroled. Patients received oral regorafenib 160 mg once daily for 3 weeks (4-week cycle). The primary endpoint was disease control rate (DCR), and secondary endpoints were safety, overall response rate (ORR), progression-free survival (PFS), and overall survival (OS). RESULTS In total, 23 patients were enrolled. c-KIT mutations were frequently reported in exon 11 (14/23, 60.9%), followed by exons 13, 17, and 9 in 5 (21.7%), 5 (21.7%), and 2 (8.7%) patients, respectively. DCR at 8 weeks was 73.9%, with 2 patients (8.7%) achieving complete response, 5 (21.7%) achieving partial response, and 10 (43.5%) showing stable disease. ORR was 30.4% (7/23). The median follow-up period was 15.7 months (95% confidence interval [CI], 9.6-21.3), and median OS and PFS were 21.5 months (95% CI, 15.1-27.9) and 7.1 months (95% CI, 5.0-9.2), respectively. Circulating tumour DNA analysis in selected patients showed high c-KIT correlation (85.7%) with tissue-based tumour mutational profiles. The most common adverse events (AEs) were skin reactions, including palmar-plantar erythrodysesthesia (52.2%), and grade 3 AEs were reported in 39.1% (9/23) of the patients. CONCLUSION Regorafenib in second- or later-line settings demonstrated significant activity in patients with metastatic melanoma harbouring c-KIT mutations.
Collapse
Affiliation(s)
- Kyoo Hyun Kim
- Division of Medical Oncology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Minkyu Jung
- Division of Medical Oncology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Hyo Jin Lee
- Department of Internal Medicine and Cancer Research Institute, Chungnam National University School of Medicine, Daejeon, Republic of Korea
| | - Su Jin Lee
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea; Divison of Hematology-Oncology, Department of Internal Medicine, Ewha Womans University College of Medicine, Seoul, Republic of Korea
| | - Miso Kim
- Department of Internal Medicine, Seoul National University Hospital, Seoul National University Cancer Research Institute, Seoul, Republic of Korea
| | - Mi Sun Ahn
- Department of Hematology-Oncology, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Moon Young Choi
- Department of Internal Medicine, Hemato-Oncology, Inje University Busan Paik Hospital, Busan, Republic of Korea
| | - Na-Ri Lee
- Division of Hematology and Oncology, Department of Internal Medicine, Jeonbuk National University Medical School, Jeonju, Republic of Korea
| | - Sang Joon Shin
- Division of Medical Oncology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
247
|
Casak SJ, Horiba MN, Yuan M, Cheng J, Lemery SJ, Shen YL, Fu W, Moore JN, Li Y, Bi Y, Auth D, Fesenko N, Kluetz PG, Pazdur R, Fashoyin-Aje LA. FDA Approval Summary: Tucatinib with Trastuzumab for Advanced Unresectable or Metastatic, Chemotherapy Refractory, HER2-Positive RAS Wild-Type Colorectal Cancer. Clin Cancer Res 2023; 29:4326-4330. [PMID: 37318379 PMCID: PMC10722550 DOI: 10.1158/1078-0432.ccr-23-1041] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 05/03/2023] [Accepted: 06/12/2023] [Indexed: 06/16/2023]
Abstract
On January 19, 2023, the FDA granted accelerated approval to tucatinib in combination with trastuzumab for the treatment of patients with unresectable or metastatic RAS wild-type, HER2-positive colorectal cancer who have received prior treatment with fluoropyrimidine, oxaliplatin, and irinotecan. Approval was based on the pooled analysis of patients receiving tucatinib in combination with trastuzumab in MOUNTAINEER (NCT03043313), an open-label, multicenter trial. The primary endpoint was overall response rate (ORR) by RECIST 1.1 as per blinded central review committee (BIRC) assessment. The main secondary endpoint was duration of response (DOR) per BIRC assessment. Eighty-four eligible patients received the combination tucatinib and trastuzumab. With a median follow-up of 16 months, the ORR was 38% [95% confidence interval (CI): 28-49] and median DOR was 12.4 months (95% CI: 8.5-20.5); 81% of responders had a response lasting more than 6 months. The most common adverse reactions observed in at least 20% of patients receiving tucatinib in combination with trastuzumab were diarrhea, fatigue, rash, nausea, abdominal pain, infusion-related reactions, and fever. FDA concluded that the magnitude of ORR and durable responses observed in patients treated with tucatinib in combination with trastuzumab in the MOUNTAINEER trial are clinically meaningful, particularly in the context of a disease with estimated survival of 6-7 months with available therapy. This is the first approval for the subset of patients with HER2-positive colorectal cancer. This article summarizes the FDA's thought process and review of the data supporting this accelerated approval.
Collapse
Affiliation(s)
- Sandra J. Casak
- Center for Drug Evaluation and Research, U.S. Food and Drug Administration
| | - M. Naomi Horiba
- Center for Drug Evaluation and Research, U.S. Food and Drug Administration
| | - Mengdie Yuan
- Center for Drug Evaluation and Research, U.S. Food and Drug Administration
| | - Joyce Cheng
- Center for Drug Evaluation and Research, U.S. Food and Drug Administration
| | - Steven J. Lemery
- Center for Drug Evaluation and Research, U.S. Food and Drug Administration
| | - Yuan Li Shen
- Center for Drug Evaluation and Research, U.S. Food and Drug Administration
| | - Wentao Fu
- Center for Drug Evaluation and Research, U.S. Food and Drug Administration
| | - Jason N. Moore
- Center for Drug Evaluation and Research, U.S. Food and Drug Administration
| | - Yangbing Li
- Center for Drug Evaluation and Research, U.S. Food and Drug Administration
| | - Youwei Bi
- Center for Drug Evaluation and Research, U.S. Food and Drug Administration
| | - Doris Auth
- Center for Drug Evaluation and Research, U.S. Food and Drug Administration
| | - Nataliya Fesenko
- Center for Drug Evaluation and Research, U.S. Food and Drug Administration
| | - Paul G. Kluetz
- Center for Drug Evaluation and Research, U.S. Food and Drug Administration
- Oncology Center of Excellence, U.S. Food and Drug Administration
| | - Richard Pazdur
- Center for Drug Evaluation and Research, U.S. Food and Drug Administration
- Oncology Center of Excellence, U.S. Food and Drug Administration
| | | |
Collapse
|
248
|
Harrelson A, Wang R, Stewart A, Ingram C, Gillis A, Rose JB, El-Rayes B, Azmi A, Chen H. Management of neuroendocrine tumor liver metastases. Am J Surg 2023; 226:623-630. [PMID: 37657968 DOI: 10.1016/j.amjsurg.2023.08.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 07/15/2023] [Accepted: 08/14/2023] [Indexed: 09/03/2023]
Abstract
BACKGROUND Neuroendocrine Tumors (NETs) are a group of tumors that arise from neuroendocrine cells, and are increasing in incidence worldwide. These tumors often metastasize to the liver, and management of these neuroendocrine tumor liver metastases (NELMs) requires a multi-disciplinary approach. We aim to provide a comprehensive update for treatment of NELMs. METHODS We completed a comprehensive systemic review of papers involving the diagnosis, treatment, and outcomes of NELMs. We identified 1612 records via Scopus database literature search. Two independent authors reviewed these records, with 318 meeting criteria for inclusion in the final systemic review. RESULTS Primary tumor resection with resection of liver metastases is the treatment of choice for patients with NELMs. Liver-directed therapies and liver transplantation can be considered for patients with unresectable liver metastases. Systemic medical therapy is used for managing tumor burden and symptoms caused by NELMs. CONCLUSIONS Advancement in liver-directed and targeted systemic therapies provide improved options for patients with unresectable tumors. Given the complexity of NELMs, management of NELMs necessitates multidisciplinary teams at comprehensive health centers.
Collapse
Affiliation(s)
- Alex Harrelson
- Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Rongzhi Wang
- Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Addison Stewart
- Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Clark Ingram
- Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Andrea Gillis
- Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, USA
| | - J Bart Rose
- Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Bassel El-Rayes
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Asfar Azmi
- Department of Oncology, Karmanos Cancer Institute, Detroit, MI, USA
| | - Herbert Chen
- Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
249
|
Ciernikova S, Sevcikova A, Drgona L, Mego M. Modulating the gut microbiota by probiotics, prebiotics, postbiotics, and fecal microbiota transplantation: An emerging trend in cancer patient care. Biochim Biophys Acta Rev Cancer 2023; 1878:188990. [PMID: 37742728 DOI: 10.1016/j.bbcan.2023.188990] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 09/13/2023] [Accepted: 09/13/2023] [Indexed: 09/26/2023]
Abstract
Treatment resistance, together with acute and late adverse effects, represents critical issues in the management of cancer patients. Promising results from preclinical and clinical research underline the emerging trend of a microbiome-based approach in oncology. Favorable bacterial species and higher gut diversity are associated with increased treatment efficacy, mainly in chemo- and immunotherapy. On the other hand, alterations in the composition and activity of gut microbial communities are linked to intestinal dysbiosis and contribute to high treatment-induced toxicity. In this Review, we provide an overview of studies concerning gut microbiota modulation in patients with solid and hematologic malignancies with a focus on probiotics, prebiotics, postbiotics, and fecal microbiota transplantation. Targeting the gut microbiome might bring clinical benefits and improve patient outcomes. However, a deeper understanding of mechanisms and large clinical trials concerning microbiome and immunological profiling is warranted to identify safe and effective ways to incorporate microbiota-based interventions in routine clinical practice.
Collapse
Affiliation(s)
- Sona Ciernikova
- Cancer Research Institute, Biomedical Research Center of Slovak Academy of Sciences, Bratislava, Slovakia.
| | - Aneta Sevcikova
- Cancer Research Institute, Biomedical Research Center of Slovak Academy of Sciences, Bratislava, Slovakia
| | - Lubos Drgona
- Department of Oncohematology, Comenius University and National Cancer Institute, Bratislava, Slovakia
| | - Michal Mego
- 2nd Department of Oncology, Comenius University and National Cancer Institute, Bratislava, Slovakia
| |
Collapse
|
250
|
Day D, Park JJ, Coward J, Markman B, Lemech C, Kuo JC, Prawira A, Brown MP, Bishnoi S, Kotasek D, Strother RM, Cosman R, Su R, Ma Y, Yue Z, Hu HH, Wu R, Li P, Tse AN. A first-in-human phase 1 study of nofazinlimab, an anti-PD-1 antibody, in advanced solid tumors and in combination with regorafenib in metastatic colorectal cancer. Br J Cancer 2023; 129:1608-1618. [PMID: 37731023 PMCID: PMC10646086 DOI: 10.1038/s41416-023-02431-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 08/29/2023] [Accepted: 09/06/2023] [Indexed: 09/22/2023] Open
Abstract
BACKGROUND We assessed nofazinlimab, an anti-PD-1 antibody, in solid tumors and combined with regorafenib in metastatic colorectal cancer (mCRC). METHODS This phase 1 study comprised nofazinlimab dose escalation (phase 1a) and expansion (phase 1b), and regorafenib dose escalation (80 or 120 mg QD, days 1-21 of 28-day cycles) combined with 300-mg nofazinlimab Q4W (part 2a) to determine safety, efficacy, and RP2D. RESULTS In phase 1a (N = 21), no dose-limiting toxicity occurred from 1 to 10 mg/kg Q3W, with 200 mg Q3W determined as the monotherapy RP2D. In phase 1b (N = 87), 400-mg Q6W and 200-mg Q3W regimens were found comparable. In part 2a (N = 14), both regimens were deemed plausible RP2Ds. Fatigue was the most frequent treatment-emergent adverse event (AE) in this study. Any-grade and grade 3/4 nofazinlimab-related AEs were 71.4% and 14.3%, 56.3% and 5.7%, and 57.1% and 21.4% in phases 1a, 1b, and part 2a, respectively. ORRs were 14.3% and 25.3% in phases 1a and 1b, respectively. In part 2a, no patients had radiological responses. CONCLUSIONS Nofazinlimab monotherapy was well tolerated and demonstrated preliminary anti-tumor activity in multiple tumor types. Regorafenib plus nofazinlimab had a manageable safety profile but was not associated with any response in mCRC. CLINICAL TRIAL REGISTR ATION Clinicaltrials.gov (NCT03475251).
Collapse
Affiliation(s)
- Daphne Day
- Department of Medical Oncology, Monash Health, Clayton, VIC, Australia.
- Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, VIC, Australia.
| | - John J Park
- Department of Clinical Medicine, Faculty of Medicine, Health and Human Sciences, Macquarie University, North Ryde, NSW, Australia
| | - Jermaine Coward
- Medical Oncology, Icon Cancer Care - South Brisbane, South Brisbane, QLD, Australia
- Faculty of Medicine, University of Queensland, Herston, QLD, Australia
| | - Ben Markman
- Department of Medical Oncology, Monash Health, Clayton, VIC, Australia
- Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, VIC, Australia
| | - Charlotte Lemech
- Drug Development, Scientia Clinical Research, Randwick, NSW, Australia
- School of Clinical Medicine, University of New South Wales, Sydney, NSW, Australia
| | - James C Kuo
- Drug Development, Scientia Clinical Research, Randwick, NSW, Australia
- School of Clinical Medicine, University of New South Wales, Sydney, NSW, Australia
| | - Amy Prawira
- School of Clinical Medicine, University of New South Wales, Sydney, NSW, Australia
- Department of Medical Oncology, The Kinghorn Cancer Centre, St. Vincent's Hospital Sydney, Darlinghurst, NSW, Australia
| | - Michael P Brown
- Cancer Clinical Trials Unit, Royal Adelaide Hospital, Adelaide, SA, Australia
- School of Medicine, University of Adelaide, Adelaide, SA, Australia
| | - Sarwan Bishnoi
- Medical Oncology, Ashford Cancer Centre Research and ICON Cancer Centre, Kurralta Park and Division of Medicine, University of Adelaide, Adelaide, SA, Australia
| | - Dusan Kotasek
- Medical Oncology, Ashford Cancer Centre Research and ICON Cancer Centre, Kurralta Park and Division of Medicine, University of Adelaide, Adelaide, SA, Australia
| | - R Matthew Strother
- Medical Oncology, Christchurch Hospital, Christchurch, Canterbury, New Zealand
| | - Rasha Cosman
- School of Clinical Medicine, University of New South Wales, Sydney, NSW, Australia
- Department of Medical Oncology, The Kinghorn Cancer Centre, St. Vincent's Hospital Sydney, Darlinghurst, NSW, Australia
| | - Rila Su
- Translational Medicine and Early Development, CStone Pharmaceuticals (Suzhou) Co., Ltd., Suzhou, China
| | - Yiding Ma
- Clinical Department, CStone Pharmaceuticals (Suzhou) Co., Ltd., Suzhou, China
| | - Zenglian Yue
- Translational Medicine and Early Development, CStone Pharmaceuticals (Suzhou) Co., Ltd., Suzhou, China
| | - Hui-Han Hu
- Translational Medicine and Early Development, CStone Pharmaceuticals (Suzhou) Co., Ltd., Suzhou, China
| | - Rachel Wu
- Clinical Department, CStone Pharmaceuticals (Suzhou) Co., Ltd., Suzhou, China
| | - Peiqi Li
- Translational Medicine and Early Development, CStone Pharmaceuticals (Suzhou) Co., Ltd., Suzhou, China
| | - Archie N Tse
- Translational Medicine and Early Development, CStone Pharmaceuticals (Suzhou) Co., Ltd., Suzhou, China
| |
Collapse
|