69 results on '"Ciombor KK"'
Search Results
2. Discovery and validation of a 10-gene predictive signature for response to adjuvant chemotherapy in stage II and III colon cancer.
- Author
-
Xu C, Xia P, Li J, Lewis KB, Ciombor KK, Wang L, Smith JJ, Beauchamp RD, and Chen XS
- Subjects
- Humans, Chemotherapy, Adjuvant methods, Prognosis, Male, Female, Gene Expression Regulation, Neoplastic drug effects, Fluorouracil therapeutic use, Fluorouracil pharmacology, Transcriptome genetics, Middle Aged, Aged, Gene Expression Profiling methods, Biomarkers, Tumor genetics, Gene Regulatory Networks, Retrospective Studies, Colonic Neoplasms genetics, Colonic Neoplasms drug therapy, Colonic Neoplasms pathology, Neoplasm Staging
- Abstract
Identifying patients with stage II and III colon cancer who will benefit from 5-fluorouracil (5-FU)-based adjuvant chemotherapy is crucial for the advancement of personalized cancer therapy. We employ a semi-supervised machine learning approach to analyze a large dataset with 933 stage II and III colon cancer samples. Our analysis leverages gene regulatory networks to discover an 18-gene prognostic signature and to explore a 10-gene signature that potentially predicts chemotherapy benefits. The 10-gene signature demonstrates strong prognostic power and shows promising potential to predict chemotherapy benefits. We establish a robust clinical assay on the NanoString nCounter platform, validated in a retrospective formalin-fixed paraffin-embedded (FFPE) cohort, which represents an important step toward clinical application. Our study lays the groundwork for improving adjuvant chemotherapy and potentially expanding into immunotherapy decision-making in colon cancer. Future prospective studies are needed to validate and establish the clinical utility of the 10-gene signature in clinical settings., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2024 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
3. NCCN Guidelines® Insights: Rectal Cancer, Version 3.2024.
- Author
-
Benson AB, Venook AP, Adam M, Chang G, Chen YJ, Ciombor KK, Cohen SA, Cooper HS, Deming D, Garrido-Laguna I, Grem JL, Haste P, Hecht JR, Hoffe S, Hunt S, Hussan H, Johung KL, Joseph N, Kirilcuk N, Krishnamurthi S, Malla M, Maratt JK, Messersmith WA, Meyerhardt J, Miller ED, Mulcahy MF, Nurkin S, Parikh A, Patel H, Pedersen K, Saltz L, Schneider C, Shibata D, Shogan B, Skibber JM, Sofocleous CT, Tavakkoli A, Willett CG, Wu C, Jones F, and Gurski L
- Subjects
- Humans, Neoadjuvant Therapy methods, Neoadjuvant Therapy standards, Combined Modality Therapy methods, Neoplasm Staging, Medical Oncology standards, Medical Oncology methods, Rectal Neoplasms therapy, Rectal Neoplasms diagnosis, Rectal Neoplasms pathology
- Abstract
The determination of an optimal treatment plan for an individual patient with rectal cancer is a complex process. In addition to decisions relating to the intent of rectal cancer surgery (ie, curative or palliative), consideration must also be given to the likely functional results of treatment, including the probability of maintaining or restoring normal bowel function/anal continence and preserving genitourinary functions. Particularly for patients with distal rectal cancer, finding a balance between curative-intent therapy while having minimal impact on quality of life can be challenging. Furthermore, the risk of pelvic recurrence is higher in patients with rectal cancer compared with those with colon cancer, and locally recurrent rectal cancer is associated with a poor prognosis. Careful patient selection and the use of sequenced multimodality therapy following a multidisciplinary approach is recommended. These NCCN Guidelines Insights detail recent updates to the NCCN Guidelines for Rectal Cancer, including the addition of endoscopic submucosal dissection as an option for early-stage rectal cancer, updates to the total neoadjuvant therapy approach based on the results of recent clinical trials, and the addition of a "watch-and-wait" nonoperative management approach for clinical complete responders to neoadjuvant therapy.
- Published
- 2024
- Full Text
- View/download PDF
4. Colon Cancer, Version 3.2024, NCCN Clinical Practice Guidelines in Oncology.
- Author
-
Benson AB, Venook AP, Adam M, Chang G, Chen YJ, Ciombor KK, Cohen SA, Cooper HS, Deming D, Garrido-Laguna I, Grem JL, Haste P, Hecht JR, Hoffe S, Hunt S, Hussan H, Johung KL, Joseph N, Kirilcuk N, Krishnamurthi S, Malla M, Maratt JK, Messersmith WA, Meyerhardt J, Miller ED, Mulcahy MF, Nurkin S, Overman MJ, Parikh A, Patel H, Pedersen K, Saltz L, Schneider C, Shibata D, Shogan B, Skibber JM, Sofocleous CT, Tavakkoli A, Willett CG, Wu C, Gurski LA, Snedeker J, and Jones F
- Subjects
- Humans, Medical Oncology standards, Medical Oncology methods, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Antineoplastic Combined Chemotherapy Protocols adverse effects, United States, Colonic Neoplasms diagnosis, Colonic Neoplasms therapy, Colonic Neoplasms pathology, Colonic Neoplasms drug therapy
- Abstract
Colorectal cancer (CRC) is the fourth most frequently diagnosed cancer and the second leading cause of cancer death in the United States. Management of disseminated metastatic CRC involves various active drugs, either in combination or as single agents. The choice of therapy is based on consideration of the goals of therapy, the type and timing of prior therapy, the mutational profile of the tumor, and the differing toxicity profiles of the constituent drugs. This manuscript summarizes the data supporting the systemic therapy options recommended for metastatic CRC in the NCCN Guidelines for Colon Cancer.
- Published
- 2024
- Full Text
- View/download PDF
5. Mutational Signature Changes in Patients With Metastatic Squamous Cell Carcinoma of the Anal Canal.
- Author
-
LaPelusa M, Cann C, Ciombor KK, and Eng C
- Subjects
- Humans, Anal Canal, Mutation, Biomarkers, Tumor genetics, High-Throughput Nucleotide Sequencing, Circulating Tumor DNA genetics, Carcinoma, Squamous Cell genetics, Anus Neoplasms genetics
- Abstract
Purpose: We examined the concordance of genetic mutations between pretreatment tumor tissue and posttreatment circulating tumor DNA (ctDNA) in patients with metastatic squamous cell carcinoma of the anal canal (SCCA) and assessed the impact of therapy on this concordance., Methods: We analyzed next-generation sequencing reports from pretreatment tumor tissue and posttreatment ctDNA in 11 patients with metastatic SCCA treated at Vanderbilt University Medical Center between 2017 and 2021., Results: Among the mutations identified in posttreatment ctDNA, 34.5% were also found in pretreatment tumor tissue, while 47.6% of pretreatment tumor tissue mutations were found in posttreatment ctDNA. Four patients had preservation of potentially actionable mutations in both pretreatment tissue and posttreatment ctDNA, while 7 patients had newly identified mutations in posttreatment ctDNA that were not present in pretreatment tumor tissue., Conclusion: Patients with SCCA demonstrate a high degree of temporal mutational heterogeneity. This supports the hypothesis that ctDNA can serve as a real-time tracking mechanism for solid tumors' molecular evolution in response to therapy. Our findings highlight the potential of ctDNA in identifying emerging actionable mutations, supplementing information from tissue-based genomic assessments. Further research, ideally with larger and multi-institutional cohorts, is needed to validate our findings in this relatively rare tumor type., (© The Author(s) 2023. Published by Oxford University Press.)
- Published
- 2024
- Full Text
- View/download PDF
6. New Opportunities for Minimizing Toxicity in Rectal Cancer Management.
- Author
-
Couwenberg AM, Varvoglis DN, Grieb BC, Marijnen CAM, Ciombor KK, and Guillem JG
- Subjects
- Humans, Immune Checkpoint Inhibitors, Immunotherapy, Chemotherapy, Adjuvant, Rectal Neoplasms, Brain Neoplasms, Neoplasms, Second Primary
- Abstract
Advances in multimodal management of locally advanced rectal cancer (LARC), consisting of preoperative chemotherapy and/or radiotherapy followed by surgery with or without adjuvant chemotherapy, have improved local disease control and patient survival but are associated with significant risk for acute and long-term morbidity. Recently published trials, evaluating treatment dose intensification via the addition of preoperative induction or consolidation chemotherapy (total neoadjuvant therapy [TNT]), have demonstrated improved tumor response rates while maintaining acceptable toxicity. In addition, TNT has led to an increased number of patients achieving a clinical complete response and thus eligible to pursue a nonoperative, organ-preserving, watch and wait approach, thereby avoiding toxicities associated with surgery, such as bowel dysfunction and stoma-related complications. Ongoing trials using immune checkpoint inhibitors in patients with mismatch repair-deficient tumors suggest that this subgroup of patients with LARC could potentially be treated with immunotherapy alone, sparing them the toxicity associated with preoperative treatment and surgery. However, the majority of rectal cancers are mismatch repair-proficient and less responsive to immune checkpoint inhibitors and require multimodal management. The synergy noted in preclinical studies between immunotherapy and radiotherapy on immunogenic tumor cell death has led to the design of ongoing clinical trials that explore the benefit of combining radiotherapy, chemotherapy, and immunotherapy (mainly of immune checkpoint inhibitors) and aim to increase the number of patients eligible for organ preservation.
- Published
- 2023
- Full Text
- View/download PDF
7. Anal Carcinoma, Version 2.2023, NCCN Clinical Practice Guidelines in Oncology.
- Author
-
Benson AB, Venook AP, Al-Hawary MM, Azad N, Chen YJ, Ciombor KK, Cohen S, Cooper HS, Deming D, Garrido-Laguna I, Grem JL, Hecht JR, Hoffe S, Hubbard J, Hunt S, Hussan H, Jeck W, Johung KL, Joseph N, Kirilcuk N, Krishnamurthi S, Maratt J, Messersmith WA, Meyerhardt J, Miller ED, Mulcahy MF, Nurkin S, Overman MJ, Parikh A, Patel H, Pedersen K, Saltz L, Schneider C, Shibata D, Skibber JM, Sofocleous CT, Stotsky-Himelfarb E, Tavakkoli A, Willett CG, Williams G, Algieri F, Gurski L, and Stehman K
- Subjects
- Humans, Biopsy, Medical Oncology, Anus Neoplasms, Carcinoma, Squamous Cell
- Abstract
This discussion summarizes the NCCN Clinical Practice Guidelines for managing squamous cell anal carcinoma, which represents the most common histologic form of the disease. A multidisciplinary approach including physicians from gastroenterology, medical oncology, surgical oncology, radiation oncology, and radiology is necessary. Primary treatment of perianal cancer and anal canal cancer are similar and include chemoradiation in most cases. Follow-up clinical evaluations are recommended for all patients with anal carcinoma because additional curative-intent treatment is possible. Biopsy-proven evidence of locally recurrent or persistent disease after primary treatment may require surgical treatment. Systemic therapy is generally recommended for extrapelvic metastatic disease. Recent updates to the NCCN Guidelines for Anal Carcinoma include staging classification updates based on the 9th edition of the AJCC Staging System and updates to the systemic therapy recommendations based on new data that better define optimal treatment of patients with metastatic anal carcinoma.
- Published
- 2023
- Full Text
- View/download PDF
8. Society for Immunotherapy of Cancer (SITC) clinical practice guideline on immunotherapy for the treatment of gastrointestinal cancer.
- Author
-
Kelly RJ, Bever K, Chao J, Ciombor KK, Eng C, Fakih M, Goyal L, Hubbard J, Iyer R, Kemberling HT, Krishnamurthi S, Ku G, Mordecai MM, Morris VK 2nd, Paulson AS, Peterson V, Shah MA, and Le DT
- Subjects
- Humans, Immunotherapy, Societies, Medical, Quality of Life, Gastrointestinal Neoplasms drug therapy
- Abstract
Gastrointestinal (GI) cancers, including esophageal, gastroesophageal junction, gastric, duodenal and distal small bowel, biliary tract, pancreatic, colon, rectal, and anal cancer, comprise a heterogeneous group of malignancies that impose a significant global burden. Immunotherapy has transformed the treatment landscape for several GI cancers, offering some patients durable responses and prolonged survival. Specifically, immune checkpoint inhibitors (ICIs) directed against programmed cell death protein 1 (PD-1), either as monotherapies or in combination regimens, have gained tissue site-specific regulatory approvals for the treatment of metastatic disease and in the resectable setting. Indications for ICIs in GI cancer, however, have differing biomarker and histology requirements depending on the anatomic site of origin. Furthermore, ICIs are associated with unique toxicity profiles compared with other systemic treatments that have long been the mainstay for GI cancer, such as chemotherapy. With the goal of improving patient care by providing guidance to the oncology community, the Society for Immunotherapy of Cancer (SITC) convened a panel of experts to develop this clinical practice guideline on immunotherapy for the treatment of GI cancer. Drawing from published data and clinical experience, the expert panel developed evidence- and consensus-based recommendations for healthcare professionals using ICIs to treat GI cancers, with topics including biomarker testing, therapy selection, and patient education and quality of life considerations, among others., Competing Interests: Competing interests: KB—researcher: Bristol-Myers Squibb, Merck. JC—consulting fees: Lilly, Merck, AstraZeneca, Foundation Medicine, Daiichi Sankyo, Macrogenics, Amgen, Ono Pharmaceutical, Bristol-Myers Squibb, Astellas, Turning Point Therapeutics, Silverback Therapeutics, Novartis, Coherus Biosciences, Geneos, Roche; fees for non-CE services: Merck, Bristol-Myers Squibb; contracted research: Merck, Brooklyn Immunotherapeutics. KKC—consulting fees: Array, Natera, Merck, Pfizer, Lilly/Loxo; contracted research: BMS, Array, Incyte, Daiichi Sankyo, Nucana, AbbVie, Merck, Pfizer/Calithera (all funds to institution). CE—consulting fees: Boston Scientific, SK, Halio Dx, J&J, Merck, Natera; contracted research: Hutchinson, Merck, Elevar, Janssen. MF—consulting fees: Amgen, Array, Bayer, fSK, HalioDx, Mirati, Pfizer, Seattle Genetics, Taiho, Zhuhai Yufan Biotech; fees for non-CE services: Guardant360, Amgen; other: Amgen, AstraZenenca, Novartis, Bristol-Myers Squibb (grants to institution). LG—consulting fees: Agios, Debiopharm, Taiho, Alentis, Incyte, Klus, Pieris, QED, SIRTEX, AstraZeneca, H3Biomedicine. JH—consulting fees: Merck, Bayer, BeiGene; contracted research: Merck, Boston Biomedical, Treos Bio, Senhwa Pharmaceuticals, Bayer, Incyte, TriOncology, Seattle Genetics, Hutchison MediPharma, Pionyr Immunotherapeutics, Trovogene, G1 Therapeutics. RI—consulting fees: Ipsen, AstraZeneca, Teresa, Hexal AG, Incyte; contracted research: Ipsen, Tersera, Merck, Aveo. RJK—researcher: Bristol-Myers Squibb and Eli Lilly with institutional grants for investigator-initiated trials awarded to Johns Hopkins and Baylor University Medical Center; consultant advisor speaker: Bristol-Myers Squibb, Ono Pharmaceuticals, Merck, AstraZeneca, Daicchi Sankyo, Astellas, Eisai, Ipsen, Pieris, Novartis, Takeda, EMD Serono, Novocure, Grail, Toray, Eli Lilly. SK—contracted research: Bristol-Myers Squibb, Aravive, Pfizer. GK—consulting fees: Apexigen, AstraZeneca, BMS, Eli Lilly, Merck, Pieris, Zymeworks; contracted research: Arog, AstraZeneca, BMS, Daiichi Sankyo, Merck, Oncolys, Pieris, Zymeworks. DTL—consulting fees: Merck, Bristol-Myers Squibb, Janssen, Nouscom; contracted research: Merck, Bristol-Myers Squibb, Aduro Biotech, Medivir, Curegenix, Nouscom; other: Merck. VKM II—consulting fees: Bicara, Servier; contracted research: Pfizer, Bristol-Myers Squibb, EMD Serono, BioNTech, Novartis. ASP—consulting fees: Helsinn, BMS, Advanced Accelerator Applications, Hutchinson, Ipsen, Incyte, Exelixis, Pfizer, QED, Lilly, Mirati, Amgen. MAS—consulting fees: Lilly Pharmaceutical; contracted research: Merck, Bristol-Myers Squibb, Oncolys Biopharma. SITC staff—CG, AK, NL, SM-W—nothing to disclose., (© Author(s) (or their employer(s)) 2023. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.)
- Published
- 2023
- Full Text
- View/download PDF
9. The Genetic Landscape of Familial Pulmonary Fibrosis.
- Author
-
Liu Q, Zhou Y, Cogan JD, Mitchell DB, Sheng Q, Zhao S, Bai Y, Ciombor KK, Sabusap CM, Malabanan MM, Markin CR, Douglas K, Ding G, Banovich NE, Nickerson DA, Blue EE, Bamshad MJ, Brown KK, Schwartz DA, Phillips JA 3rd, Martinez-Barricarte R, Salisbury ML, Shyr Y, Loyd JE, Kropski JA, and Blackwell TS
- Subjects
- Humans, Endothelial Cells, Risk Factors, Telomere, Genetic Predisposition to Disease genetics, Receptors, Lysophosphatidic Acid genetics, Pulmonary Fibrosis genetics, Lung Diseases, Interstitial genetics
- Abstract
Rationale and Objectives: Up to 20% of idiopathic interstitial lung disease is familial, referred to as familial pulmonary fibrosis (FPF). An integrated analysis of FPF genetic risk was performed by comprehensively evaluating for genetic rare variants (RVs) in a large cohort of FPF kindreds. Methods: Whole-exome sequencing and/or candidate gene sequencing from affected individuals in 569 FPF kindreds was performed, followed by cosegregation analysis in large kindreds, gene burden analysis, gene-based risk scoring, cell-type enrichment analysis, and coexpression network construction. Measurements and Main Results: It was found that 14.9-23.4% of genetic risk in kindreds could be explained by RVs in genes previously linked to FPF, predominantly telomere-related genes. New candidate genes were identified in a small number of families-including SYDE1 , SERPINB8 , GPR87 , and NETO1 -and tools were developed for evaluation and prioritization of RV-containing genes across kindreds. Several pathways were enriched for RV-containing genes in FPF, including focal adhesion and mitochondrial complex I assembly. By combining single-cell transcriptomics with prioritized candidate genes, expression of RV-containing genes was discovered to be enriched in smooth muscle cells, type II alveolar epithelial cells, and endothelial cells. Conclusions: In the most comprehensive FPF genetic study to date, the prevalence of RVs in known FPF-related genes was defined, and new candidate genes and pathways relevant to FPF were identified. However, new RV-containing genes shared across multiple kindreds were not identified, thereby suggesting that heterogeneous genetic variants involving a variety of genes and pathways mediate genetic risk in most FPF kindreds.
- Published
- 2023
- Full Text
- View/download PDF
10. Tucatinib plus trastuzumab for chemotherapy-refractory, HER2-positive, RAS wild-type unresectable or metastatic colorectal cancer (MOUNTAINEER): a multicentre, open-label, phase 2 study.
- Author
-
Strickler JH, Cercek A, Siena S, André T, Ng K, Van Cutsem E, Wu C, Paulson AS, Hubbard JM, Coveler AL, Fountzilas C, Kardosh A, Kasi PM, Lenz HJ, Ciombor KK, Elez E, Bajor DL, Cremolini C, Sanchez F, Stecher M, Feng W, and Bekaii-Saab TS
- Subjects
- Humans, Male, Female, Middle Aged, Trastuzumab adverse effects, Receptor, ErbB-2 genetics, Cohort Studies, Antibodies, Monoclonal, Humanized adverse effects, Diarrhea chemically induced, Antineoplastic Combined Chemotherapy Protocols adverse effects, Colonic Neoplasms drug therapy, Rectal Neoplasms
- Abstract
Background: HER2 is an actionable target in metastatic colorectal cancer. We assessed the activity of tucatinib plus trastuzumab in patients with chemotherapy-refractory, HER2-positive, RAS wild-type unresectable or metastatic colorectal cancer., Methods: MOUNTAINEER is a global, open-label, phase 2 study that enrolled patients aged 18 years and older with chemotherapy-refractory, HER2-positive, RAS wild-type unresectable or metastatic colorectal cancer at 34 sites (clinics and hospitals) in five countries (Belgium, France, Italy, Spain, and the USA). Initially, the study was designed as a single-cohort study, which was expanded following an interim analysis to include more patients. Initially, patients were given tucatinib (300 mg orally twice daily) plus intravenous trastuzumab (8 mg/kg as an initial loading dose, then 6 mg/kg every 21 days; cohort A) for the duration of treatment (until progression), and after expansion, patients were randomly assigned (4:3), using an interactive web response system and stratified by primary tumour location, to either tucatinib plus trastuzumab (cohort B) or tucatinib monotherapy (cohort C). The primary endpoint was confirmed objective response rate per blinded independent central review (BICR) for cohorts A and B combined and was assessed in patients in the full analysis set (ie, patients with HER2-positive disease who received at least one dose of study treatment). Safety was assessed in all patients who received at least one dose of study treatment. This trial is registered with ClinicalTrials.gov, NCT03043313, and is ongoing., Findings: Between Aug 8, 2017, and Sept 22, 2021, 117 patients were enrolled (45 in cohort A, 41 in cohort B, and 31 in cohort C), of whom 114 patients had locally assessed HER2-positive disease and received treatment (45 in cohort A, 39 in cohort B, and 30 in cohort C; full analysis set), and 116 patients received at least one dose of study treatment (45 in cohort A, 41 in cohort B, and 30 in cohort C; safety population). In the full analysis set, median age was 56·0 years (IQR 47-64), 66 (58%) were male, 48 (42%) were female, 88 (77%) were White, and six (5%) were Black or African American. As of data cutoff (March 28, 2022), in 84 patients from cohorts A and B in the full analysis set, the confirmed objective response rate per BICR was 38·1% (95% CI 27·7-49·3; three patients had a complete response and 29 had a partial response). In cohorts A and B, the most common adverse event was diarrhoea (55 [64%] of 86), the most common grade 3 or worse adverse event was hypertension (six [7%] of 86), and three (3%) patients had tucatinib-related serious adverse events (acute kidney injury, colitis, and fatigue). In cohort C, the most common adverse event was diarrhoea (ten [33%] of 30), the most common grade 3 or worse adverse events were increased alanine aminotransferase and aspartate aminotransferase (both two [7%]), and one (3%) patient had a tucatinib-related serious adverse event (overdose). No deaths were attributed to adverse events. All deaths in treated patients were due to disease progression., Interpretation: Tucatinib plus trastuzumab had clinically meaningful anti-tumour activity and favourable tolerability. This treatment is the first US Food and Drug Administration-approved anti-HER2 regimen for metastatic colorectal cancer and is an important new treatment option for chemotherapy-refractory HER2-positive metastatic colorectal cancer., Funding: Seagen and Merck & Co., Competing Interests: Declaration of interests JHS reports support from Seagen for the present manuscript; grants paid to the institution by Amgen, Bayer, Erasca, Seagen, Daiichi-Sankyo, Gossamer Bio, AStar D3, Sanofi, Roche/Genentech, Curegenix, Nektar, AbbVie, and Silverback Therapeutics; receiving consulting fees from AbbVie, Takeda, AstraZeneca, Bayer, GlaxoSmithKline, Inivata, Mereo Biopharma, Pfizer, Roche/Genentech, Seagen, Silverback Therapeutics, and Viatris; receiving honoraria from Bayer, Natera, and Pfizer; receiving travel support from Seagen and Guardant Health; receiving other services from Guardant Health; and is a member of advisory boards for AbbVie and Pionyr Immunotherapeutics. AC reports grants paid to their institution by Seagen, GlaxoSmithKline, and Inspira (previously RGenix) and receiving advisory or consultancy fees from Bayer, Merck, Seagen, GlaxoSmithKline, Janssen, and G1 Therapeutics. SS is a member of advisory boards for Agenus, AstraZeneca, Bayer, Bristol Myers Squibb, CheckmAb, Daiichi-Sankyo, Guardant Health, Menarini, Merck, Novartis, Pierre-Fabre, Roche-Genentech, and Seagen. TA reports attending advisory board meetings and receiving consulting fees from Astellas, Aptitude Health, Bristol Myers Squibb, Gritstone Oncology, GamaMabs Pharma Sa, GlaxoSmithKline, Gilead, Kaleido Oncology, Merck & Co, Pierre Fabre, Seagen, Servier Transgène, and Roche/Ventana; receiving support for attending meetings from Bristol Myers Squibb, Merck & Co, and Servier; and receiving honoraria for speaking, manuscript writing, or educational events from AstraZeneca, Bristol Myers Squibb, Gritstone Oncology, GlaxoSmithKline, Merck & Co, Pierre Fabre, Seagen, Roche/Ventana, Sanofi, and Servier; and receiving travel support from Bristol Myers Squibb and Merck & Co. KN reports grants from Pharmavite, Revolution Medicines, Evergrande Group, and Janssen and receiving consultancy fees from Bayer, Seagen, X-Biotix Therapeutics, Array Biopharma, BiomX, Bicara Therapeutics, GlaxoSmithKline, Pfizer, and Redesign Health. EVC reports grants paid to the institution by Amgen, Bayer, Boehringer Ingelheim, Bristol-Myers Squibb, Celgene, Ipsen, Lilly, Merck Sharp & Dohme, Merck KGaA, Novartis, Roche, and Servier and receiving consulting fees from AbbVie, Array, Astellas, AstraZeneca, Bayer, Beigene, Biocartis, Boehringer Ingelheim, Bristol Myers Squibb, Celgene, Daiichi, Halozyme, GlaxoSmithKline, Helsinn, Incyte, Ipsen, Janssen Research, Lilly, Merck Sharp & Dohme, Merck KGaA, Mirati, Novartis, Pierre Fabre, Roche, Seattle Genetics, Servier, Sirtex, Terumo, Taiho, TRIGR, and Zymeworks. CW reports grants paid to the institution by Boston Biomedical, Bristol Myers Squibb, Lycera, RAPT Therapeutics, Seagen, Symphogan, Vaccinex, INHBRX, and Pfizer; receiving honoraria from Array Biopharma, Signatera, Pfizer, Daiichi Sankyo; receiving travel support from Array Biopharma; and former employment at Winship Cancer Institute of Emory University, Atlanta, GA, USA (at the time of study conduct). ASP reports receiving support for the present from Seagen to provide medical writing assistance, institutional funding, study materials, processing charges; funding paid to the institution for clinical research trials from Ipsen, Bristol Myers Squibb, Exelixis, HutchMed, Seagen, Taiho, Lilly, AstraZeneca, Incyte, Deciphera, G1 Therapeutics, Zentalis, Tempus, Camurus, Relay Therapeutics, Nucana, Merck, and Bayer; receiving honoraria from Ideo Oncology and MJH Life Sciences (both educational event companies); participating in advisory boards for Amgen, Bristol Myers Squibb, Eisai, Ipsen, AAA, Exelixis, Pfizer, QED, Lilly, Mirati, HutchMed, Astellas, and Aadi; receiving travel support from Pfizer; owning stock or stock options from Aptose, Actinium, and Alexion; and receiving medical writing services from Bayer, Ipsen, HutchMed, Exelixis, and Seagen. JMH reports grants paid to the institution by Merck, Boston Biomedical, Senhwa Pharmaceuticals, Bayer, Incyte, TriOncology, Seattle Genetics, Hutchison MediPharma, Pionyr Immunotherapeutics, Trovogene, G1 Therapeutics, Roche, and Treos Bio and participation on advisory boards with honoraria to the institution by Bayer, Merck, BeiGene, and Incyte. ALC reports grants from Nucana, Seagen, Abgenomics, Novocure, Actuate, Medimmune/AstraZeneca, PanCan, Amgen, and Nextrast. CF reports support for the present manuscript paid to the institution by Seattle Genetics (currently Seagen) and grants paid to their institution by the National Comprehensive Cancer Network Foundation, National Comprehensive Cancer Network Oncology Research Program, Taiho Oncology, National Cancer Institute (NCI), and Pfizer. AK reports no conflict of interest. PMK reports grants paid to the institution by Advanced Accelerator Applications, Tersera, and Boston Scientific; a consultancy and advisory board relationship with Elicio (scientific advisory board member/shares/stock ownership); receiving consulting fees from Natera, Foundation Medicine, Illumina, BostonGene, Merck/MSD Oncology, Tempus, Bayer, Lilly, Delcath Systems, IPBA, QED Therapeutics, Boston Healthcare Associates, Servier, Taiho Oncology, Exact Sciences, Daiichi Sankyo/AstraZeneca, Eisai, and Seattle Genetics; consulting fees paid to the institution by Taiho Pharmaceutical and Ipsen; receiving travel support from AstraZeneca; and former employment at University of Iowa Hospitals and Clinics, Iowa City, IA, USA (at the time of study conduct). H-JL reports grants from NCI (P30, U01, U2c, UG1, and UM1), US Department of Defense, Southwest Oncology Group; receiving consulting fees from Merck, Bayer, Merck KG, Roche, Fulgent, Oncocyte, G1 Therapeutics, Jazz Therapeutics, 3T bioscience, and Adagene; receiving honoraria from Bayer, Roche, and Pfizer; participating in unpaid leadership roles with Wunderglo Foundation, Debbie Dream Foundation, and Cancer Support Group; and owning stock options in Fulgent. KKC reports grants paid to the institution by Bristol Myers Squibb, Array, Incyte, Daiichi Sankyo, Nucana, Merck, Pfizer, Calithera, Genentech, and Seagen; participating on advisory boards and receiving consulting fees from Merck, Lilly/Loxo, Pfizer, Replimune, Personalis, Exelixis, and Array; receiving travel or hotel support for attending advisory board meeting from Array; and participating on the Board of Directors for Academic and Community Cancer Research United. EE reports grants, receiving consulting fees and payment for expert testimony, receiving honoraria, receiving travel support, and participating in advisory boards from Amgen, Bayer, Hoffmann-La Roche, Merck Serono, Sanofi, Pierre Fabre, MSD, Organon, Novartis, Servier and financial support for clinical trials or contracted research paid to the institution from Amgen Inc, Array Biopharma Inc, AstraZeneca Pharmaceuticals LP, BeiGene, Boehringer Ingelheim, Bristol Myers Squibb, Celgene, Debiopharm International SA, F Hoffmann-La Roche, Genentech, HalioDX SAS, Hutchison MediPharma International, Janssen-Cilag SA, MedImmune, Menarini, Merck Health KGAA, Merck Sharp & Dohme, Merus NV, Mirati, Novartis Farmaceutica SA, Pfizer, Pharma Mar, Sanofi Aventis Recherche & Developpement, Servier, Taiho Pharma USA. DLB reports a grant for trial support of the present study paid to the institution by Seagen; grants paid to the institution by Seagen, Rafael/Cornerstone, and Abbvie; receiving honoraria for speakers bureau from Natera; receiving salary support for participating on the local data safety monitoring board for University Hospitals; receiving no payment for participating on the advisory board for Rafael pharmaceuticals; and receiving study drug from Novartis and Epizyme. CC reports grants paid to the institution by Merck, Bayer, Servier, Amgen, and Roche; receiving honoraria from Roche, Amgen, Merck, MSD, Pierre Fabre, Servier, Bayer, and Nordic Pharma; receiving payment for expert testimony from Merck and Bayer; receiving travel support from Amgen; participating on advisory boards for Nordic Pharma, Amgen, MSD, Pierre Fabre, and Mirati. FS reports no conflict of interest. MS reports being employed by and owning stock in Seagen. WF reports being employed by and owning stock in Seagen. TSB-S reports research funding paid to the institution by Agios, Arys, Arcus, Atreca, Boston Biomedical, Bayer, Eisai, Celgene, Lilly, Ipsen, Clovis, Seattle Genetics, Genentech, Novartis, Mirati, Merus, Abgenomics, Incyte, Pfizer, and Bristol Myers Squibb; consulting fees paid to the institution by Ipsen, Arcus, Pfizer, Seattle Genetics, Bayer, Genentech, Incyte, Eisai, and Merck; receiving consulting fees from Stemline, AbbVie, Boehringer Ingelheim, Janssen, Daichii Sankyo, Natera, TreosBio, Celularity, Exact Science, Sobi, Beigene, Kanaph, Astra Zeneca, Deciphera, MJH Life Sciences, Aptitude Health, Illumina, and Foundation Medicine; participating on independent data monitoring committees or data safety monitoring boards for Fibrogen, Suzhou Kintor, AstraZeneca, Exelixis, Merck/Eisai, PanCan, and 1Globe; participating on scientific advisory boards for Imugene, Immuneering, Xilis, Replimune Artiva, and Sun Biopharma; receiving royalties from Uptodate; and inventions or patents—WO/2018/183488: HUMAN PD1 PEPTIDE VACCINES AND USES THEREOF (licensed to Imugene) and WO/2019/055687: METHODS AND COMPOSITIONS FOR THE TREATMENT OF CANCER CACHEXIA (licensed to Recursion)., (Copyright © 2023 Elsevier Ltd. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
11. Immunotherapy in Localized Microsatellite Instability-High/Mismatch Repair Deficient Solid Tumors: Are We Ready for a New Standard of Care?
- Author
-
Ciombor KK and Eng C
- Subjects
- Humans, DNA Mismatch Repair, Standard of Care, Immunotherapy, Microsatellite Instability, Colorectal Neoplasms
- Published
- 2023
- Full Text
- View/download PDF
12. Treatment of Metastatic Colorectal Cancer: ASCO Guideline.
- Author
-
Morris VK, Kennedy EB, Baxter NN, Benson AB 3rd, Cercek A, Cho M, Ciombor KK, Cremolini C, Davis A, Deming DA, Fakih MG, Gholami S, Hong TS, Jaiyesimi I, Klute K, Lieu C, Sanoff H, Strickler JH, White S, Willis JA, and Eng C
- Subjects
- Humans, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Cetuximab, Endothelial Growth Factors therapeutic use, Practice Guidelines as Topic, Colonic Neoplasms drug therapy, Colorectal Neoplasms therapy, Colorectal Neoplasms drug therapy, Rectal Neoplasms drug therapy
- Abstract
Purpose: To develop recommendations for treatment of patients with metastatic colorectal cancer (mCRC)., Methods: ASCO convened an Expert Panel to conduct a systematic review of relevant studies and develop recommendations for clinical practice., Results: Five systematic reviews and 10 randomized controlled trials met the systematic review inclusion criteria., Recommendations: Doublet chemotherapy should be offered, or triplet therapy may be offered to patients with previously untreated, initially unresectable mCRC, on the basis of included studies of chemotherapy in combination with anti-vascular endothelial growth factor antibodies. In the first-line setting, pembrolizumab is recommended for patients with mCRC and microsatellite instability-high or deficient mismatch repair tumors; chemotherapy and anti-epidermal growth factor receptor therapy is recommended for microsatellite stable or proficient mismatch repair left-sided treatment-naive RAS wild-type mCRC; chemotherapy and anti-vascular endothelial growth factor therapy is recommended for microsatellite stable or proficient mismatch repair RAS wild-type right-sided mCRC. Encorafenib plus cetuximab is recommended for patients with previously treated BRAF V600E-mutant mCRC that has progressed after at least one previous line of therapy. Cytoreductive surgery plus systemic chemotherapy may be recommended for selected patients with colorectal peritoneal metastases; however, the addition of hyperthermic intraperitoneal chemotherapy is not recommended. Stereotactic body radiation therapy may be recommended following systemic therapy for patients with oligometastases of the liver who are not considered candidates for resection. Selective internal radiation therapy is not routinely recommended for patients with unilobar or bilobar metastases of the liver. Perioperative chemotherapy or surgery alone should be offered to patients with mCRC who are candidates for potentially curative resection of liver metastases. Multidisciplinary team management and shared decision making are recommended. Qualifying statements with further details related to implementation of guideline recommendations are also included.Additional information is available at www.asco.org/gastrointestinal-cancer-guidelines.
- Published
- 2023
- Full Text
- View/download PDF
13. Dietary fat in relation to all-cause mortality and cancer progression and death among people with metastatic colorectal cancer: Data from CALGB 80405 (Alliance)/SWOG 80405.
- Author
-
Van Blarigan EL, Ma C, Ou FS, Bainter TM, Venook AP, Ng K, Niedzwiecki D, Giovannucci E, Lenz HJ, Polite BN, Hochster HS, Goldberg RM, Mayer RJ, Blanke CD, O'Reilly EM, Ciombor KK, and Meyerhardt JA
- Subjects
- Female, Animals, Male, Dietary Fats, Diet, Cause of Death, Cardiovascular Diseases, Colonic Neoplasms, Rectal Neoplasms
- Abstract
Data on diet and survival among people with metastatic colorectal cancer are limited. We examined dietary fat in relation to all-cause mortality and cancer progression or death among 1149 people in the Cancer and Leukemia Group B (Alliance)/Southwest Oncology Group (SWOG) 80405 trial who completed a food frequency questionnaire at initiation of treatment for advanced or metastatic colorectal cancer. We examined saturated, monounsaturated, total and specific types (n-3, long-chain n-3 and n-6) of polyunsaturated fat, animal and vegetable fats. We hypothesized higher vegetable fat intake would be associated with lower risk of all-cause mortality and cancer progression. We used Cox proportional hazards regression to estimate adjusted hazard ratios (HR) and 95% confidence intervals (CI). Over median follow-up of 6.1 years (interquartile range [IQR]: 5.3, 7.2 y), we observed 974 deaths and 1077 events of progression or death. Participants had a median age of 59 y; 41% were female and 86% identified as White. Moderate or higher vegetable fat was associated with lower risk of mortality and cancer progression or death (HRs comparing second, third and fourth to first quartile for all-cause mortality: 0.74 [0.62, 0.90]; 0.75 [0.61, 0.91]; 0.79 [0.63, 1.00]; P trend: .12; for cancer progression or death: 0.74 [0.62, 0.89]; 0.78 [0.64, 0.95]; 0.71 [0.57, 0.88]; P trend: .01). No other fat type was associated with all-cause mortality and cancer progression or death. Moderate or higher vegetable fat intake may be associated with lower risk of cancer progression or death among people with metastatic colorectal cancer., (© 2022 UICC.)
- Published
- 2023
- Full Text
- View/download PDF
14. Rectal Cancer, Version 2.2022, NCCN Clinical Practice Guidelines in Oncology.
- Author
-
Benson AB, Venook AP, Al-Hawary MM, Azad N, Chen YJ, Ciombor KK, Cohen S, Cooper HS, Deming D, Garrido-Laguna I, Grem JL, Gunn A, Hecht JR, Hoffe S, Hubbard J, Hunt S, Jeck W, Johung KL, Kirilcuk N, Krishnamurthi S, Maratt JK, Messersmith WA, Meyerhardt J, Miller ED, Mulcahy MF, Nurkin S, Overman MJ, Parikh A, Patel H, Pedersen K, Saltz L, Schneider C, Shibata D, Skibber JM, Sofocleous CT, Stotsky-Himelfarb E, Tavakkoli A, Willett CG, Gregory K, and Gurski L
- Subjects
- Humans, Medical Oncology, Neoadjuvant Therapy, Rectal Neoplasms diagnosis, Rectal Neoplasms pathology, Rectal Neoplasms therapy
- Abstract
This selection from the NCCN Guidelines for Rectal Cancer focuses on management of malignant polyps and resectable nonmetastatic rectal cancer because important updates have been made to these guidelines. These recent updates include redrawing the algorithms for stage II and III disease to reflect new data supporting the increasingly prominent role of total neoadjuvant therapy, expanded recommendations for short-course radiation therapy techniques, and new recommendations for a "watch-and-wait" nonoperative management technique for patients with cancer that shows a complete response to neoadjuvant therapy. The complete version of the NCCN Guidelines for Rectal Cancer, available online at NCCN.org, covers additional topics including risk assessment, pathology and staging, management of metastatic disease, posttreatment surveillance, treatment of recurrent disease, and survivorship.
- Published
- 2022
- Full Text
- View/download PDF
15. Diagnosis and Management of Rectal Cancer in Patients Younger Than 50 Years: Rising Global Incidence and Unique Challenges.
- Author
-
Lang D and Ciombor KK
- Subjects
- Cohort Studies, Humans, Incidence, Adenocarcinoma pathology, Colorectal Neoplasms diagnosis, Rectal Neoplasms diagnosis, Rectal Neoplasms epidemiology, Rectal Neoplasms therapy
- Abstract
The global incidence of colorectal adenocarcinoma is stable or decreasing overall; however, the incidence of colorectal cancer in patients aged <50 years is increasing. Although some of this increase is due to hereditary cancer syndromes, this is not the sole explanation. Patients with early-onset rectal cancer in particular have unique disease patterns and face distinct challenges in their treatment. Molecular patterns of disease in this patient cohort are noteworthy and often represent an opportunity to target these cancers more effectively. Recent and ongoing trials focusing on minimizing toxicities and necessary therapy modalities and maximizing response and patient outcome are of paramount importance in this patient population. Additional resources are needed for this patient population, including fertility counseling and preservation, financial guidance, genetic counseling, and psychosocial support.
- Published
- 2022
- Full Text
- View/download PDF
16. Anal Cancer: Emerging Standards in a Rare Disease.
- Author
-
Eng C, Ciombor KK, Cho M, Dorth JA, Rajdev LN, Horowitz DP, Gollub MJ, Jácome AA, Lockney NA, Muldoon RL, Washington MK, O'Brian BA, Benny A, Lebeck Lee CM, Benson AB 3rd, Goodman KA, and Morris VK
- Subjects
- Anal Canal pathology, Humans, Rare Diseases complications, Rare Diseases pathology, Anus Neoplasms diagnosis, Anus Neoplasms epidemiology, Anus Neoplasms therapy, Carcinoma, Squamous Cell diagnosis, Carcinoma, Squamous Cell epidemiology, Carcinoma, Squamous Cell therapy, HIV Infections epidemiology, Papillomavirus Infections
- Abstract
The social stigma surrounding an anal cancer diagnosis has traditionally prevented open discussions about this disease. However, as recent treatment options and an increasing rate of diagnoses are made worldwide, awareness is growing. In the United States alone, 9,090 individuals were expected to be diagnosed with anal cancer in 2021. The US annual incidence of squamous cell carcinoma of the anus continues to increase by 2.7% yearly, whereas the mortality rate increases by 3.1%. The main risk factor for anal cancer is a human papillomavirus infection; those with chronic immunosuppression are also at risk. Patients with HIV are 19 times more likely to develop anal cancer compared with the general population. In this review, we have provided an overview of the carcinoma of the anal canal, the role of screening, advancements in radiation therapy, and current trials investigating acute and chronic treatment-related toxicities. This article is a comprehensive approach to presenting the existing data in an effort to encourage continuous international interest in anal cancer.
- Published
- 2022
- Full Text
- View/download PDF
17. BRAF -Mutated Advanced Colorectal Cancer: A Rapidly Changing Therapeutic Landscape.
- Author
-
Ciombor KK, Strickler JH, Bekaii-Saab TS, and Yaeger R
- Subjects
- Antineoplastic Combined Chemotherapy Protocols therapeutic use, Cetuximab, Humans, Mutation, Protein Kinase Inhibitors pharmacology, Protein Kinase Inhibitors therapeutic use, Colorectal Neoplasms drug therapy, Colorectal Neoplasms genetics, Colorectal Neoplasms pathology, Proto-Oncogene Proteins B-raf genetics
- Abstract
BRAF -mutated advanced colorectal cancer is a relatively small but critical subset of this tumor type on the basis of prognostic and predictive implications. BRAF alterations in colorectal cancer are classified into three functional categories on the basis of signaling mechanisms, with the class I BRAF
V600E mutation occurring most frequently in colorectal cancer. Functional categorization of BRAF mutations in colorectal cancer demonstrates distinct mitogen-activated protein kinase pathway signaling. On the basis of recent clinical trials, current standard-of-care therapies for patients with BRAFV600E - mutated metastatic colorectal cancer include first-line cytotoxic chemotherapy plus bevacizumab and subsequent therapy with the BRAF inhibitor encorafenib and antiepidermal growth factor receptor antibody cetuximab. Treatment regimens currently under exploration in BRAFV600E -mutant metastatic colorectal cancer include combinatorial options of various pathway-targeted therapies, cytotoxic chemotherapy, and/or immune checkpoint blockade, among others. Circumvention of adaptive and acquired resistance to BRAF-targeted therapies is a significant challenge to be overcome in BRAF -mutated advanced colorectal cancer.- Published
- 2022
- Full Text
- View/download PDF
18. A contemporary systematic review on liver transplantation for unresectable liver metastases of colorectal cancer.
- Author
-
Lebeck Lee CM, Ziogas IA, Agarwal R, Alexopoulos SP, Ciombor KK, Matsuoka LK, Brown DB, and Eng C
- Subjects
- Hepatectomy, Humans, Prospective Studies, Retrospective Studies, Colorectal Neoplasms pathology, Colorectal Neoplasms surgery, Liver Neoplasms secondary, Liver Neoplasms surgery, Liver Transplantation
- Abstract
The 5-year overall survival rate of a patient with unresectable metastatic colorectal cancer is poor at approximately 14%. Similarly, historical data on liver transplantation (LT) in those with colorectal liver metastases (CRLM) showed poor outcomes, with 5-year survival rates between 12% and 21%. More recently, limited data have shown improved outcomes in select patients with 5-year overall survival rates of approximately 60%. Despite these reported survival improvements, there is no significant improvement in disease-free survival. Given the uncertain benefit with this therapeutic approach and a renewed investigational interest, we aimed to conduct a contemporary systematic review on LT for CRLM. A systematic review of the literature was performed according to the preferred reporting items for systematic reviews and meta-analysis statement. English articles reporting on data regarding LT for CRLM were identified through the MEDLINE (via PubMed), Cochrane Library, and ClinicalTrials.gov databases (last search date: December 16th, 2021) by 2 researchers independently. A total of 58 studies (45 published and 13 ongoing) were included. Although early retrospective studies suggest the possibility that some carefully selected patients may benefit from LT, there is minimal prospective data on the topic and LT remains exploratory in the setting of CRLM. Additionally, several other challenges, such as the limited availability of deceased donor organs and defining appropriate selection criteria, remain when considering the implementation of LT for these patients. Further evidence from ongoing prospective trials is needed to determine if and to what extent there is a role for LT in patients with surgically unresectable CRLM., (© 2022 The Authors. Cancer published by Wiley Periodicals LLC on behalf of American Cancer Society.)
- Published
- 2022
- Full Text
- View/download PDF
19. Systemic therapy for gastric cancer: Perioperative strategies and beyond.
- Author
-
Cann C and Ciombor KK
- Subjects
- Antineoplastic Combined Chemotherapy Protocols therapeutic use, Chemotherapy, Adjuvant, Gastrectomy, Humans, Neoadjuvant Therapy, Stomach Neoplasms drug therapy, Stomach Neoplasms surgery
- Abstract
Each year, gastric cancer claims the lives of hundreds of thousands of patients worldwide. Despite surgical resection, the risk of residual disease, micrometastatic disease, and disease recurrence remain elevated. Herein, we review systemic therapy strategies in the neoadjuvant, adjuvant, and metastatic settings, including novel uses of immunotherapy, targeted therapies and cytotoxic chemotherapies, for the treatment of gastric cancer., (© 2022 Wiley Periodicals LLC.)
- Published
- 2022
- Full Text
- View/download PDF
20. Immunotherapy for Microsatellite Stable Colorectal Cancers: Challenges and Novel Therapeutic Avenues.
- Author
-
Sahin IH, Ciombor KK, Diaz LA, Yu J, and Kim R
- Subjects
- DNA Mismatch Repair, Humans, Immune Checkpoint Inhibitors, Immunotherapy, Microsatellite Repeats, Tumor Microenvironment genetics, Colorectal Neoplasms drug therapy, Colorectal Neoplasms therapy, Microsatellite Instability
- Abstract
With the development of immune checkpoint inhibitors, immunotherapy researchers have facilitated substantial progress for patients with mismatch repair deficient/microsatellite instability-high colorectal cancer, which has led to practice changes at a head-spinning pace. However, this benefit has not been translated into microsatellite stable colorectal cancer, which carries the hallmarks of chromosomal instability. So far, clinical trials have not shown any substantial clinical benefits of immune checkpoint inhibitor therapy for patients with microsatellite stable colorectal cancer, which has been disappointing. Recently, combinations of immune checkpoint inhibitors with tyrosine kinase inhibitors and targeted therapies have been investigated for potential synergistic effects that may increase antitumor activity in the tumor microenvironment and achieve more substantial clinical and radiologic responses. In this article, we discuss the current state of the science for the use of immune checkpoint inhibitors in microsatellite stable colorectal cancers, and we review the molecular underpinnings of inherited physiologic barriers for the delivery of effective immunotherapy. We also elaborate on existing therapeutic opportunities to convert microsatellite stable colorectal cancer into an "immune hot" cancer, which may define the future treatment paradigm of colorectal cancer for which there is a great unmet need.
- Published
- 2022
- Full Text
- View/download PDF
21. External Validation of a Clinical Score for Patients With Neuroendocrine Tumors Under Consideration for Peptide Receptor Radionuclide Therapy.
- Author
-
Das S, Chauhan A, Du L, Thomas KE, Jacob A, Schad A, Jain S, Jessop A, Shah C, Eisner D, Cardin DB, Ciombor KK, Goff LW, Bradshaw M, Delbeke D, Sandler M, Ramirez RA, and Berlin J
- Subjects
- Aged, Cohort Studies, Female, Humans, Kaplan-Meier Estimate, Male, Middle Aged, Octreotide analogs & derivatives, Octreotide therapeutic use, Organometallic Compounds therapeutic use, Predictive Value of Tests, Prognosis, Progression-Free Survival, Proportional Hazards Models, Prospective Studies, Radionuclide Imaging, Treatment Outcome, Neuroendocrine Tumors mortality, Neuroendocrine Tumors radiotherapy, Radioisotopes therapeutic use, Receptors, Peptide therapeutic use, Severity of Illness Index
- Abstract
Importance: Despite the benefit of peptide receptor radionuclide therapy (PRRT) for patients with well-differentiated neuroendocrine tumors (WD NETs), no clinical metric to anticipate benefit from the therapy for individual patients has been previously defined., Objective: To assess whether the prognostic ability of the clinical score (CS) could be validated in an external cohort of patients with WD NETs., Design, Setting, and Participants: This multicenter cohort study's analysis included patients with WD NETs who were under consideration for peptide receptor radionuclide therapy (PRRT) with lutetium-177 (177Lu)-dotatate between March 1, 2016, and March 17, 2020. The original cohort included patients from Vanderbilt-Ingram Cancer Center. The validation cohort included patients from Ochsner Medical Center, Markey Cancer Center, and Rush Medical Center. Patients with paragangliomas, pheochromocytomas and neuroblastomas were excluded. Statistical analysis was performed from June to November 2021., Exposures: PRRT with 177Lu-dotatate or alternate therapies such as everolimus, sunitinib, or capecitabine plus temozolomide., Main Outcomes and Measures: The primary outcome was progression-free survival (PFS) and was estimated by the Kaplan-Meier method; a Cox proportional-hazards model adjusting for primary tumor site, tumor grade, and number of PRRT doses administered was used to analyze association between CS and outcomes., Results: A total of 126 patients (median age [IQR] age: 63.6 [52.9-70.7] years; 64 male individuals) were included in the validation cohort, and the combined cohort (validation and original cohorts combined) had a total of 248 patients (median [IQR] patient age: 63.3 [53.3-70.3] years; 126 male individuals). In the validation cohort, on multivariable analysis, for each 2-point increase in CS, PFS decreased significantly (hazard ratio, 2.61; 95% CI, 1.64-4.16). After finding an association of the CS with PFS in the validation cohort, the original and validation cohorts were combined into the cohort for this analysis. On multivariable analysis, for each 2-point increase in CS, PFS decreased significantly (hazard ratio, 2.52; 95% CI, 1.89-3.36)., Conclusions and Relevance: Increases in CS were associated with worsening PFS in the validation cohort, validating findings from the original cohort. These findings suggest that the CS, to our knowledge, represents the first clinical metric to estimate anticipated benefit from PRRT for patients with WD NETs and may be a clinical tool for patients being considered for PRRT.
- Published
- 2022
- Full Text
- View/download PDF
22. First-in-Human PET Imaging and Estimated Radiation Dosimetry of l-[5- 11 C]-Glutamine in Patients with Metastatic Colorectal Cancer.
- Author
-
Cohen AS, Grudzinski J, Smith GT, Peterson TE, Whisenant JG, Hickman TL, Ciombor KK, Cardin D, Eng C, Goff LW, Das S, Coffey RJ, Berlin JD, and Manning HC
- Subjects
- Humans, Male, Female, Middle Aged, Aged, Positron-Emission Tomography, Tissue Distribution, Positron Emission Tomography Computed Tomography, Carbon Radioisotopes, Radiopharmaceuticals pharmacokinetics, Colorectal Neoplasms diagnostic imaging, Colorectal Neoplasms metabolism, Colorectal Neoplasms pathology, Glutamine metabolism, Radiometry, Neoplasm Metastasis
- Abstract
Altered metabolism is a hallmark of cancer. In addition to glucose, glutamine is an important nutrient for cellular growth and proliferation. Noninvasive imaging via PET may help facilitate precision treatment of cancer through patient selection and monitoring of treatment response. l-[5-
11 C]-glutamine (11 C-glutamine) is a PET tracer designed to study glutamine uptake and metabolism. The aim of this first-in-human study was to evaluate the radiologic safety and biodistribution of11 C-glutamine for oncologic PET imaging. Methods: Nine patients with confirmed metastatic colorectal cancer underwent PET/CT imaging. Patients received 337.97 ± 44.08 MBq of11 C-glutamine. Dynamic PET acquisitions that were centered over the abdomen or thorax were initiated simultaneously with intravenous tracer administration. After the dynamic acquisition, a whole-body PET/CT scan was acquired. Volume-of-interest analyses were performed to obtain estimates of organ-based absorbed doses of radiation. Results:11 C-glutamine was well tolerated in all patients, with no observed safety concerns. The organs with the highest radiation exposure included the bladder, pancreas, and liver. The estimated effective dose was 4.46E-03 ± 7.67E-04 mSv/MBq. Accumulation of11 C-glutamine was elevated and visualized in lung, brain, bone, and liver metastases, suggesting utility for cancer imaging. Conclusion: PET using11 C-glutamine appears safe for human use and allows noninvasive visualization of metastatic colon cancer lesions in multiple organs. Further studies are needed to elucidate its potential for other cancers and for monitoring response to treatment., (© 2022 by the Society of Nuclear Medicine and Molecular Imaging.)- Published
- 2022
- Full Text
- View/download PDF
23. Combining tumor deposits with the number of lymph node metastases to improve the prognostic accuracy in stage III colon cancer: a post hoc analysis of the CALGB/SWOG 80702 phase III study (Alliance) ☆ .
- Author
-
Cohen R, Shi Q, Meyers J, Jin Z, Svrcek M, Fuchs C, Couture F, Kuebler P, Ciombor KK, Bendell J, De Jesus-Acosta A, Kumar P, Lewis D, Tan B, Bertagnolli MM, Philip P, Blanke C, O'Reilly EM, Shields A, and Meyerhardt JA
- Subjects
- Humans, Lymph Nodes pathology, Lymphatic Metastasis, Neoplasm Staging, Prognosis, Retrospective Studies, Colonic Neoplasms pathology, Extranodal Extension
- Abstract
Background: In colon cancer, tumor deposits (TD) are considered in assigning prognosis and staging only in the absence of lymph node metastasis (i.e. stage III pN1c tumors). We aimed to evaluate the prognostic value of the presence and the number of TD in patients with stage III, node-positive colon cancer., Patients and Methods: All participants from the CALGB/SWOG 80702 phase III trial were included in this post hoc analysis. Pathology reports were reviewed for the presence and the number of TD, lymphovascular and perineural invasion. Associations with disease-free survival (DFS) and overall survival (OS) were evaluated by multivariable Cox models adjusting for sex, treatment arm, T-stage, N-stage, lymphovascular invasion, perineural invasion and lymph node ratio., Results: Overall, 2028 patients were included with 524 (26%) TD-positive and 1504 (74%) TD-negative tumors. Of the TD-positive patients, 80 (15.4%) were node negative (i.e. pN1c), 239 (46.1%) were pN1a/b (<4 positive lymph nodes) and 200 (38.5%) were pN2 (≥4 positive lymph nodes). The presence of TD was associated with poorer DFS [adjusted hazard ratio (aHR) = 1.63, 95% CI 1.33-1.98] and OS (aHR = 1.59, 95% CI 1.24-2.04). The negative effect of TD was observed for both pN1a/b and pN2 groups. Among TD-positive patients, the number of TD had a linear negative effect on DFS and OS. Combining TD and the number of lymph node metastases, 104 of 1470 (7.1%) pN1 patients were re-staged as pN2, with worse outcomes than patients confirmed as pN1 (3-year DFS rate: 65.4% versus 80.5%, P = 0.0003; 5-year OS rate: 87.9% versus 69.1%, P = <0.0001). DFS was not different between patients re-staged as pN2 and those initially staged as pN2 (3-year DFS rate: 65.4% versus 62.3%, P = 0.4895)., Conclusion: Combining the number of TD and the number of lymph node metastases improved the prognostication accuracy of tumor-node-metastasis (TNM) staging., Competing Interests: Disclosure RC declares honoraria from MSD Oncology and Servier, and research grants from the ARCAD foundation, the Nuovo-Soldati foundation, the ARC foundation for cancer research and from the Servier Institute. All other authors have declared no conflicts of interest., (Copyright © 2021 European Society for Medical Oncology. Published by Elsevier Ltd. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
24. Mismatch Repair (MMR) Gene Alteration and BRAF V600E Mutation Are Potential Predictive Biomarkers of Immune Checkpoint Inhibitors in MMR-Deficient Colorectal Cancer.
- Author
-
Sahin IH, Goyal S, Pumpalova Y, Sonbol MB, Das S, Haraldsdottir S, Ahn D, Ciombor KK, Chen Z, Draper A, Berlin J, Bekaii-Saab T, Lesinski GB, El-Rayes BF, and Wu C
- Subjects
- Aged, Biomarkers, Biomarkers, Tumor genetics, DNA Mismatch Repair genetics, Humans, Immune Checkpoint Inhibitors, Microsatellite Instability, MutL Protein Homolog 1 metabolism, Mutation, Proto-Oncogene Proteins B-raf genetics, Colonic Neoplasms, Colorectal Neoplasms drug therapy, Colorectal Neoplasms genetics
- Abstract
Background: Immune checkpoint inhibitor (ICI) therapy is highly effective in metastatic mismatch repair-deficient (MMR-D) colorectal cancer (CRC). In this study, we evaluated molecular and clinical predictors of ICI response in MMR-D CRC., Materials and Methods: Patient databases at four cancer institutions were queried. The Fisher exact test was performed to test the association of clinical and molecular markers. The Kaplan-Meier method was used to estimate progression-free survival (PFS) and compared by the log-rank test. Twelve- and 24-month PFS rates were compared by the Z test., Results: A total of 60 patients with CRC with MMR-D/microsatellite instability-high who previously received ICIs were identified. Patients with liver metastasis had a lower overall response rate as compared with other sites of metastasis (36.4% vs. 68.7%; p = .081). Patients with MLH1/PMS2 loss had worse 1-year and 2-year PFS rates compared with patients with MSH2/MSH6 loss (84.2% vs. 57.8% and 78.2% vs. 54.2%, respectively; p < .001). There were improved 1-year and 2-year PFS rates in patients with wild-type BRAF when compared with patients with BRAF V600E mutation (73.3% vs. 40%, and 73.3% vs. 26.7%; respectively; p < .001). Patients aged >65 had significantly worse PFS rates as compared with patients aged ≤65 (p < .001)., Conclusion: BRAF V600E mutation, MLH1 and/or PMS2 loss, as well as age >65 years and liver metastasis, may be predictive of duration of ICI response in patients with MMR-D CRC. Larger cohorts are needed to confirm our findings., Implications for Practice: The results of this study reveal clinically important biomarkers that potentially predict immune checkpoint inhibitor response in patients with mismatch repair-deficient colorectal cancer., (© 2021 AlphaMed Press.)
- Published
- 2021
- Full Text
- View/download PDF
25. Safety considerations with new treatment regimens for anal cancer.
- Author
-
Cimino SK, Ciombor KK, Chakravarthy AB, Bailey CE, Hopkins MB, Geiger TM, Hawkins AT, and Eng C
- Subjects
- Anus Neoplasms epidemiology, Anus Neoplasms pathology, Humans, Incidence, Salvage Therapy methods, Anus Neoplasms therapy, Chemoradiotherapy methods, Immunotherapy methods
- Abstract
Introduction : Anal cancer is a rare malignancy, but incidence rates are rising. Primary chemoradiation is the standard of care for early disease with surgery reserved for salvage. Despite success in terms of survival, patients suffer significant morbidity. Research is underway to advance the field and improve outcomes for these patients. Areas covered : This review aims to discuss the safety and efficacy of new approaches to treat anal cancer. A literature search was performed from January 1950 through November 2020 via PubMed and ClinicalTrials.gov databases to obtain data from ongoing or published studies examining new regimens for the treatment of anal cancers. Pertinent topics covered include miniature drug conjugates, epidermal growth factor receptor inhibitors, checkpoint inhibitor combinations, and novel immunomodulators. Expert opinion : Based on emerging clinical data, the treatment paradigm for anal cancer is likely to shift in the upcoming years. One of the largest areas of investigation is the field of immunotherapy, which may emerge as an integral component of anal cancer for all treatment settings.
- Published
- 2021
- Full Text
- View/download PDF
26. Surgical resection and survival outcomes in metastatic young adult colorectal cancer patients.
- Author
-
Arhin ND, Shen C, Bailey CE, Matsuoka LK, Hawkins AT, Holowatyj AN, Ciombor KK, Hopkins MB, Geiger TM, Kam AE, Roth MT, Lebeck Lee CM, Lapelusa M, Dasari A, and Eng C
- Subjects
- Adult, Colorectal Neoplasms epidemiology, Colorectal Neoplasms pathology, Female, Humans, Incidence, Kaplan-Meier Estimate, Male, Metastasectomy statistics & numerical data, Palliative Care, Proportional Hazards Models, SEER Program, Time Factors, Young Adult, Colorectal Neoplasms mortality, Colorectal Neoplasms surgery, Metastasectomy mortality
- Abstract
Background: The incidence of colorectal cancer in adults younger than age 50 has increased with rates expected to continue to increase over the next decade. The objective of this study is to examine the survival benefit of surgical resection (primary and/or metastatic) versus palliative therapy in this patient population., Methods: We identified 6708 young adults aged 18-45 years diagnosed with metastatic colorectal cancer (mCRC) from 2004 to 2015 from the SEER database. Overall survival (OS) was analyzed using Kaplan-Meier estimation, log rank test, and multivariate Cox proportional hazards model., Results: Sixty-three percent of patients in our study underwent primary tumor resection (PTR), with 40% undergoing PTR alone and 23% undergoing both resection of primary disease and metastasectomy. The median OS for patients who underwent both PTR and metastasectomy was 36 months, compared to 13 months for those who did not receive any surgical intervention. The multivariate analysis showed significant OS benefit of receiving both PTR and metastasectomy (HR 0.34, 95% CI: 0.31-0.37, p < 0.001) compared to palliative therapy. Undergoing PTR only and metastasectomy only were also associated with improved OS (HR 0.46, 95% CI: 0.43-0.49, p < 0.001 and HR 0.64, 95% CI: 0.55-0.76, p < 0.001, respectively)., Conclusion: This is the largest observational study to evaluate survival outcomes in young-onset mCRC patients and the role of surgical intervention of the primary and/or metastatic site. Our study provides evidence of statistically significant increase in OS for young mCRC patients who undergo surgical intervention of the primary and/or metastatic site., (© 2021 The Authors. Cancer Medicine published by John Wiley & Sons Ltd.)
- Published
- 2021
- Full Text
- View/download PDF
27. Preemptive Versus Reactive Topical Clobetasol for Regorafenib-Induced Hand-Foot Reactions: A Preplanned Analysis of the ReDOS Trial.
- Author
-
Jatoi A, Ou FS, Ahn DH, Zemla TJ, Le-Rademacher JG, Boland P, Ciombor KK, Jacobs NL, Pasche B, Cleary JM, McCune JS, Pedersen KS, Barzi A, Chiorean EG, Heying EN, Lenz HJ, Sloan JA, Grothey A, Lacouture ME, and Bekaii-Saab T
- Subjects
- Activities of Daily Living, Humans, Phenylurea Compounds, Pyridines, Quality of Life, Clobetasol therapeutic use, Hand-Foot Syndrome drug therapy, Hand-Foot Syndrome etiology, Hand-Foot Syndrome prevention & control
- Abstract
Background: Hand-foot skin reaction (HFSR) is the most common regorafenib-induced adverse event and is in need of effective prevention and palliation., Materials and Methods: The Regorafenib Dose Optimization Study (ReDOS), a four-arm, previously published trial with a 1:1:1:1 randomization scheme, was analyzed in a manner in keeping with the original protocol to assess whether clobetasol 0.05% cream (a corticosteroid) applied to the palms and soles twice per day for 8 weeks was more effective when prescribed preemptively (before the development of HFSR) versus reactively (after the development of HFSR). Patients were assessed during the first two cycles of regorafenib., Results: Sixty-one patients received preemptive clobetasol, and 55 received reactive clobetasol. Groups were balanced on demographics. Over the first two cycles, no evidence of HFSR occurred in 30% with preemptive clobetasol versus 13% with reactive clobetasol (p = .03). During the first cycle, 54% and 45% of patients had no HFSR with preemptive and reactive clobetasol, respectively (p = .35). During the second cycle, 33% and 15% had no HFSR with preemptive and reactive clobetasol, respectively (p = .02). During the second cycle, rates of grade 1, 2, and 3 HFSR were 30%, 8%, and 3%, respectively, with preemptive clobetasol and 43%, 18%, and 7%, respectively, with reactive clobetasol (p = .12). Patient-reported outcomes showed HFSR compromised nearly all activities of daily living with worse quality of life in patients who received reactive versus preemptive clobetasol. No clobetasol-induced adverse events were reported., Conclusion: Preemptive clobetasol might lessen regorafenib-induced hand-foot reactions compared with reactive therapy. Further confirmatory studies are needed in a larger patient cohort., Implications for Practice: Regorafenib causes hand-foot skin reactions. Preemptive clobetasol, a high-potency topical corticosteroid, appears to lessen the severity of this adverse event. Although further study is needed, the favorable adverse event profile of this intervention might prompt clinicians to discuss this option with their patients., (© 2021 AlphaMed Press.)
- Published
- 2021
- Full Text
- View/download PDF
28. Rectothecal Fistula Secondary to a Tailgut Cyst With Malignant Transformation: An Abnormal Connection and Unusual Pathology.
- Author
-
Stewart MK, Kavalukas SL, Bonfield CM, Ciombor KK, Shi C, Kachnic LA, and Hawkins AT
- Subjects
- Adenocarcinoma diagnosis, Adult, Cerebral Ventriculitis etiology, Cysts surgery, Diagnosis, Differential, Female, Humans, Meningitis etiology, Rectal Diseases surgery, Rectal Fistula complications, Rectal Fistula surgery, Rectal Neoplasms diagnosis, Cell Transformation, Neoplastic, Cysts complications, Rectal Diseases complications, Rectal Fistula etiology, Rectal Fistula pathology
- Published
- 2021
- Full Text
- View/download PDF
29. Colon Cancer, Version 2.2021, NCCN Clinical Practice Guidelines in Oncology.
- Author
-
Benson AB, Venook AP, Al-Hawary MM, Arain MA, Chen YJ, Ciombor KK, Cohen S, Cooper HS, Deming D, Farkas L, Garrido-Laguna I, Grem JL, Gunn A, Hecht JR, Hoffe S, Hubbard J, Hunt S, Johung KL, Kirilcuk N, Krishnamurthi S, Messersmith WA, Meyerhardt J, Miller ED, Mulcahy MF, Nurkin S, Overman MJ, Parikh A, Patel H, Pedersen K, Saltz L, Schneider C, Shibata D, Skibber JM, Sofocleous CT, Stoffel EM, Stotsky-Himelfarb E, Willett CG, Gregory KM, and Gurski LA
- Subjects
- Biosimilar Pharmaceuticals, DNA Mismatch Repair, Humans, Microsatellite Instability, Mutation, Colonic Neoplasms diagnosis, Colonic Neoplasms genetics, Colonic Neoplasms therapy
- Abstract
This selection from the NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines) for Colon Cancer focuses on systemic therapy options for the treatment of metastatic colorectal cancer (mCRC), because important updates have recently been made to this section. These updates include recommendations for first-line use of checkpoint inhibitors for mCRC, that is deficient mismatch repair/microsatellite instability-high, recommendations related to the use of biosimilars, and expanded recommendations for biomarker testing. The systemic therapy recommendations now include targeted therapy options for patients with mCRC that is HER2-amplified, or BRAF V600E mutation-positive. Treatment and management of nonmetastatic or resectable/ablatable metastatic disease are discussed in the complete version of the NCCN Guidelines for Colon Cancer available at NCCN.org. Additional topics covered in the complete version include risk assessment, staging, pathology, posttreatment surveillance, and survivorship.
- Published
- 2021
- Full Text
- View/download PDF
30. Immune-Related Adverse Events and Immune Checkpoint Inhibitor Efficacy in Patients with Gastrointestinal Cancer with Food and Drug Administration-Approved Indications for Immunotherapy.
- Author
-
Das S, Ciombor KK, Haraldsdottir S, Pumpalova Y, Sahin IH, Pineda G, Shyr Y, Lin EP, Hsu CY, Chu SK, Goff LW, Cardin DB, Bilen MA, Fisher GA, Wu C, and Berlin J
- Subjects
- Humans, Immunotherapy adverse effects, Prospective Studies, Retrospective Studies, United States, United States Food and Drug Administration, Gastrointestinal Neoplasms drug therapy, Immune Checkpoint Inhibitors
- Abstract
Introduction: Immune-related adverse event (IRAE) onset may represent a clinical biomarker for anti-programmed cell death protein 1 (PD-1) antibody response based on emerging evidence from patients with various advanced malignancies. This phenomenon has not been previously reported in a multidisease cohort of patients with gastrointestinal (GI) cancer with Food and Drug Administration (FDA)-approved indications to receive immune checkpoint inhibitor therapy., Materials and Methods: The study was a multicenter retrospective cohort analysis of 76 patients with GI cancer who had received anti-PD-1 antibodies for FDA-approved indications. The primary and secondary outcomes of the study were progression-free survival (PFS) and overall survival (OS) in patients based upon IRAE presence, respectively. PFS and OS were estimated by the Kaplan-Meier method; a Cox proportional-hazards model adjusted for IRAE onset, patient age, and enrolling institution was used to analyze outcomes., Results: Median PFS and OS were prolonged in patients who experienced IRAEs compared with those who did not experience them (PFS: not reached [NR] vs. 3.9 months [hazard ratio (HR) 0.13, 95% confidence interval (CI) 0.05-0.3, p < .001]; OS: NR vs. 7.4 months [HR 0.11, 95% CI 0.03-0.36, p < .001]). Among patients who experienced IRAEs, there were no significant differences in PFS and OS by either initial IRAE severity, management, or time to onset., Conclusion: Patients with gastrointestinal cancer who experienced IRAEs while on anti-PD-1 antibodies demonstrated significant improvements in PFS and OS compared with their counterparts who did not develop IRAEs. Although these findings add to results from studies in other tumor types, larger prospective studies are needed prior to clinical adoption of IRAE onset as a biomarker for immune checkpoint inhibitor response., Implications for Practice: Predictive clinical biomarkers for immune checkpoint inhibitor response have been understudied in the field of immuno-oncology. Immune-related adverse event onset appears to be one such biomarker. Across tumor types, immune-related adverse event onset has been associated with response to anti-programmed cell death protein 1 (PD-1) antibodies. The results of this study demonstrate this for the first time in patients with gastrointestinal cancer receiving anti-PD-1 antibodies. Before immune-related adverse event onset can be adopted clinically as a predictive biomarker for immune checkpoint inhibitor response, however, larger prospective studies are needed to better understand the nuances between immune-related adverse event characteristics (severity, site, management, timing of onset) and immune checkpoint inhibitor effectiveness., (© AlphaMed Press 2020.)
- Published
- 2020
- Full Text
- View/download PDF
31. Phase I Trial of Trametinib with Neoadjuvant Chemoradiation in Patients with Locally Advanced Rectal Cancer.
- Author
-
Wu C, Williams TM, Robb R, Webb A, Wei L, Chen W, Mikhail S, Ciombor KK, Cardin DB, Timmers C, Krishna SG, Arnold M, Harzman A, Abdel-Misih S, Roychowdhury S, Bekaii-Saab T, and Wuthrick E
- Subjects
- Aged, Biomarkers, Tumor, Chemoradiotherapy, Combined Modality Therapy, Disease Management, Female, Humans, Kaplan-Meier Estimate, Male, Middle Aged, Molecular Targeted Therapy, Neoadjuvant Therapy, Neoplasm Grading, Neoplasm Staging, Prognosis, Protein Kinase Inhibitors administration & dosage, Protein Kinase Inhibitors adverse effects, Pyridones administration & dosage, Pyridones adverse effects, Pyrimidinones administration & dosage, Pyrimidinones adverse effects, Rectal Neoplasms etiology, Rectal Neoplasms metabolism, Treatment Outcome, Protein Kinase Inhibitors therapeutic use, Pyridones therapeutic use, Pyrimidinones therapeutic use, Rectal Neoplasms pathology, Rectal Neoplasms therapy
- Abstract
Purpose: The RAS/RAF/MEK/ERK signaling pathway is critical to the development of colorectal cancers, and KRAS , NRAS , and BRAF mutations foster resistance to radiation. We performed a phase I trial to determine the safety of trametinib, a potent MEK1/2 inhibitor, with 5-fluorouracil (5-FU) chemoradiation therapy (CRT) in patients with locally advanced rectal cancer (LARC)., Patients and Methods: Patients with stage II/III rectal cancer were enrolled on a phase I study with 3+3 study design, with an expansion cohort of 9 patients at the MTD. Following a 5-day trametinib lead-in, with pre- and posttreatment tumor biopsies, patients received trametinib and CRT, surgery, and adjuvant chemotherapy. Trametinib was given orally daily at 3 dose levels: 0.5 mg, 1 mg, and 2 mg. CRT consisted of infusional 5-FU 225 mg/m
2 /day and radiation dose of 28 daily fractions of 1.8 Gy (total 50.4 Gy). The primary endpoint was to identify the MTD and recommended phase II dose. IHC staining for phosphorylated ERK (pERK) and genomic profiling was performed on the tumor samples., Results: Patients were enrolled to all dose levels, and 18 patients were evaluable for toxicities and responses. Treatment was well tolerated, and there was one dose-limiting toxicity of diarrhea, which was attributed to CRT rather than trametinib. At the 2 mg dose level, 25% had pathologic complete response. IHC staining confirmed dose-dependent decrease in pERK with increasing trametinib doses., Conclusions: The combination of trametinib with 5-FU CRT is safe and well tolerated, and may warrant additional study in a phase II trial, perhaps in a RAS/RAF -mutant selected population., (©2020 American Association for Cancer Research.)- Published
- 2020
- Full Text
- View/download PDF
32. NCCN Guidelines Insights: Rectal Cancer, Version 6.2020.
- Author
-
Benson AB, Venook AP, Al-Hawary MM, Arain MA, Chen YJ, Ciombor KK, Cohen S, Cooper HS, Deming D, Garrido-Laguna I, Grem JL, Gunn A, Hoffe S, Hubbard J, Hunt S, Kirilcuk N, Krishnamurthi S, Messersmith WA, Meyerhardt J, Miller ED, Mulcahy MF, Nurkin S, Overman MJ, Parikh A, Patel H, Pedersen K, Saltz L, Schneider C, Shibata D, Skibber JM, Sofocleous CT, Stoffel EM, Stotsky-Himelfarb E, Willett CG, Johnson-Chilla A, and Gurski LA
- Subjects
- Humans, Neoadjuvant Therapy, Practice Guidelines as Topic, Colonic Neoplasms diagnosis, Colonic Neoplasms therapy, Rectal Neoplasms diagnosis, Rectal Neoplasms therapy
- Abstract
The NCCN Guidelines for Rectal Cancer provide recommendations for the diagnosis, evaluation, treatment, and follow-up of patients with rectal cancer. These NCCN Guidelines Insights summarize the panel discussion behind recent important updates to the guidelines. These updates include clarifying the definition of rectum and differentiating the rectum from the sigmoid colon; the total neoadjuvant therapy approach for localized rectal cancer; and biomarker-targeted therapy for metastatic colorectal cancer, with a focus on new treatment options for patients with BRAF V600E- or HER2 amplification-positive disease.
- Published
- 2020
- Full Text
- View/download PDF
33. Efficacy of FGFR Inhibitors and Combination Therapies for Acquired Resistance in FGFR2-Fusion Cholangiocarcinoma.
- Author
-
Krook MA, Lenyo A, Wilberding M, Barker H, Dantuono M, Bailey KM, Chen HZ, Reeser JW, Wing MR, Miya J, Samorodnitsky E, Smith AM, Dao T, Martin DM, Ciombor KK, Hays J, Freud AG, and Roychowdhury S
- Subjects
- Apoptosis, Bile Duct Neoplasms genetics, Bile Duct Neoplasms metabolism, Bile Duct Neoplasms pathology, Biomarkers, Tumor genetics, Cell Proliferation, Cholangiocarcinoma genetics, Cholangiocarcinoma metabolism, Cholangiocarcinoma pathology, Female, Gene Expression Regulation, Neoplastic, Humans, Middle Aged, Mutation, Prognosis, Receptor, Fibroblast Growth Factor, Type 2 genetics, Signal Transduction, Tumor Cells, Cultured, Antineoplastic Combined Chemotherapy Protocols pharmacology, Bile Duct Neoplasms drug therapy, Biomarkers, Tumor metabolism, Cholangiocarcinoma drug therapy, Drug Resistance, Neoplasm, Oncogene Proteins, Fusion genetics, Phenylurea Compounds therapeutic use, Protein Kinase Inhibitors therapeutic use, Pyrimidines therapeutic use, Receptor, Fibroblast Growth Factor, Type 2 antagonists & inhibitors
- Abstract
The fibroblast growth factor receptor (FGFR) signaling pathway is aberrantly activated in approximately 15% to 20% of patients with intrahepatic cholangiocarcinoma. Currently, several FGFR kinase inhibitors are being assessed in clinical trials for patients with FGFR-altered cholangiocarcinoma. Despite evidence of initial responses and disease control, virtually all patients eventually develop acquired resistance. Thus, there is a critical need for the development of innovative therapeutic strategies to overcome acquired drug resistance. Here, we present findings from a patient with FGFR2-altered metastatic cholangiocarcinoma who enrolled in a phase II clinical trial of the FGFR inhibitor, infigratinib (BGJ398). Treatment was initially effective as demonstrated by imaging and tumor marker response; however, after 8 months on trial, the patient exhibited tumor regrowth and disease progression. Targeted sequencing of tumor DNA after disease progression revealed the FGFR2 kinase domain p.E565A and p.L617M single-nucleotide variants (SNV) hypothesized to drive acquired resistance to infigratinib. The sensitivities of these FGFR2 SNVs, which were detected post-infigratinib therapy, were extended to include clinically relevant FGFR inhibitors, including AZD4547, erdafitinib (JNJ-42756493), dovitinib, ponatinib, and TAS120, and were evaluated in vitro Through a proteomics approach, we identified upregulation of the PI3K/AKT/mTOR signaling pathway in cells harboring the FGFR2 p.E565A mutation and demonstrated that combination therapy strategies with FGFR and mTOR inhibitors may be used to overcome resistance to FGFR inhibition, specific to infigratinib. Collectively, these studies support the development of novel combination therapeutic strategies in addition to the next generation of FGFR inhibitors to overcome acquired resistance in patients., (©2020 American Association for Cancer Research.)
- Published
- 2020
- Full Text
- View/download PDF
34. Early detection of SARS-CoV-2 from staging PET-CT.
- Author
-
Khattab MH, Sherry AD, Jessop AC, Ciombor KK, and Chakravarthy B
- Abstract
Objective: SARS-CoV-2 infection may manifest with minimal or no clinical symptoms. However, signs of infection may appear on routine imaging obtained in the care of patients with cancer. The management of patients planned for chemoradiation with asymptomatic or mildly symptomatic SARS-CoV-2 infection is uncertain., Methods: Here, we present a case study of a mildly symptomatic patient with anal cancer diagnosed with SARS-CoV-2 from a staging PET-CT scan., Results: PET-CT scan for anal cancer staging demonstrated pulmonary avidity suspicious for an infectious, rather than malignant, process. In the setting of these imaging findings and new-onset anosmia, viral polymerase chain reaction was ordered and found to be positive for SARS-CoV-2. To avoid myelosuppression in the setting of active infection, planned chemoradiation was delayed until cessation of viral shedding., Conclusion: In the COVID-19 era, oncologists obtaining routine staging imaging should have high diagnostic suspicion for subclinical SARS-CoV-2 infection. To avoid precipitating severe pneumonia and hospitalization, multidisciplinary discussion with risk-benefit analysis is recommended before initiating immunosuppressive therapies such as chemoradiation., Competing Interests: Conflict of interestThe authors declare that they have no conflict of interest., (© Springer-Verlag GmbH Germany, part of Springer Nature 2020.)
- Published
- 2020
- Full Text
- View/download PDF
35. Small Bowel Adenocarcinoma, Version 1.2020, NCCN Clinical Practice Guidelines in Oncology.
- Author
-
Benson AB, Venook AP, Al-Hawary MM, Arain MA, Chen YJ, Ciombor KK, Cohen SA, Cooper HS, Deming DA, Garrido-Laguna I, Grem JL, Hoffe SE, Hubbard J, Hunt S, Kamel A, Kirilcuk N, Krishnamurthi S, Messersmith WA, Meyerhardt J, Miller ED, Mulcahy MF, Nurkin S, Overman MJ, Parikh A, Patel H, Pedersen KS, Saltz LB, Schneider C, Shibata D, Skibber JM, Sofocleous CT, Stoffel EM, Stotsky-Himelfarb E, Willett CG, Johnson-Chilla A, Gregory KM, and Gurski LA
- Subjects
- Adenocarcinoma etiology, Adenocarcinoma mortality, Combined Modality Therapy, Diagnosis, Differential, Humans, Intestinal Neoplasms etiology, Intestinal Neoplasms mortality, Neoplasm Staging, Risk Factors, Survivorship, Treatment Outcome, Watchful Waiting, Adenocarcinoma diagnosis, Adenocarcinoma therapy, Intestinal Neoplasms diagnosis, Intestinal Neoplasms therapy, Intestine, Small pathology, Practice Guidelines as Topic
- Abstract
Small bowel adenocarcinoma (SBA) is a rare malignancy of the gastrointestinal tract that has increased in incidence across recent years. Often diagnosed at an advanced stage, outcomes for SBA are worse on average than for other related malignancies, including colorectal cancer. Due to the rarity of this disease, few studies have been done to direct optimal treatment, although recent data have shown that SBA responds to treatment differently than colorectal cancer, necessitating a separate approach to treatment. The NCCN Guidelines for Small Bowel Adenocarcinoma were created to establish an evidence-based standard of care for patients with SBA. These guidelines provide recommendations on the workup of suspected SBA, primary treatment options, adjuvant treatment, surveillance, and systemic therapy for metastatic disease. Additionally, principles of imaging and endoscopy, pathologic review, surgery, radiation therapy, and survivorship are described.
- Published
- 2019
- Full Text
- View/download PDF
36. Regorafenib dose-optimisation in patients with refractory metastatic colorectal cancer (ReDOS): a randomised, multicentre, open-label, phase 2 study.
- Author
-
Bekaii-Saab TS, Ou FS, Ahn DH, Boland PM, Ciombor KK, Heying EN, Dockter TJ, Jacobs NL, Pasche BC, Cleary JM, Meyers JP, Desnoyers RJ, McCune JS, Pedersen K, Barzi A, Chiorean EG, Sloan J, Lacouture ME, Lenz HJ, and Grothey A
- Subjects
- Adult, Aged, Antineoplastic Agents adverse effects, Dose-Response Relationship, Drug, Female, Humans, Male, Middle Aged, Phenylurea Compounds adverse effects, Pyridines adverse effects, Adenocarcinoma drug therapy, Antineoplastic Agents administration & dosage, Colorectal Neoplasms drug therapy, Phenylurea Compounds administration & dosage, Pyridines administration & dosage
- Abstract
Background: Regorafenib confers an overall survival benefit in patients with refractory metastatic colorectal cancer; however, the adverse event profile of regorafenib has limited its use. Despite no supportive evidence, various dosing schedules are used clinically to alleviate toxicities. This study evaluated the safety and activity of two regorafenib dosing schedules., Methods: In this randomised, multicentre, open-label, phase 2 study done in 39 outpatient cancer centres in the USA, adults aged 18 years or older with histologically or cytologically confirmed advanced or metastatic adenocarcinoma of the colon or rectum that was refractory to previous standard therapy, including EGFR inhibitors if KRAS wild-type, were enrolled. Eligible patients had an Eastern Cooperative Oncology Group performance status of 0-1 and had no previous treatment with regorafenib. Patients were randomly assigned (1:1:1:1) into four groups with two distinct regorafenib dosing strategies and two clobetasol usage plans, stratified by hospital. Regorafenib dosing strategies were a dose-escalation strategy (starting dose 80 mg/day orally with weekly escalation, per 40 mg increment, to 160 mg/day regorafenib) if no significant drug-related adverse events occurred and a standard-dose strategy (160 mg/day orally) for 21 days of a 28-day cycle. Clobetasol usage plans (0·05% clobetasol cream twice daily applied to palms and soles) were either pre-emptive or reactive. After randomisation to the four preplanned groups, using the Pocock and Simon dynamic allocation procedures stratified by the treating hospitals, we formally tested the interaction between the two interventions, dosing strategy and clobetasol usage. Given the absence of a significant interaction (p=0·74), we decided to pool the data for the pre-emptive and reactive treatment with clobetasol and compared the two dosing strategies (dose escalation vs standard dose). The primary endpoint was the proportion of evaluable patients (defined as those who were eligible, consented, and received any protocol treatment) initiating cycle 3 and was analysed per protocol. Superiority for dose escalation was declared if the one-sided p value with Fisher's exact test was less than 0·2. This trial is registered with ClinicalTrials.gov, number NCT02368886. This study is fully accrued but remains active., Findings: Between June 2, 2015, and June 22, 2017, 123 patients were randomly assigned to treatment, of whom 116 (94%) were evaluable. The per-protocol population consisted of 54 patients in the dose-escalation group and 62 in the standard-dose group. At data cutoff on July 24, 2018, median follow-up was 1·18 years (IQR 0·98-1·57). The primary endpoint was met: 23 (43%, 95% CI 29-56) of 54 patients in the dose-escalation group initiated cycle 3 versus 16 (26%, 15-37) of 62 patients in the standard-dose group (one-sided p=0·043). The most common grade 3-4 adverse events were fatigue (seven [13%] patients in the dose-escalation group vs 11 [18%] in the standard-dose group), hand-foot skin reaction (eight [15%] patients vs ten [16%] patients), abdominal pain (nine [17%] patients vs four [6%] patients), and hypertension (four [7%] patients vs nine [15%] patients). 14 patients had at least one drug-related serious adverse event: six patients in the dose-escalation group and eight patients in the standard-dose group. There was one probable treatment-related death in the standard-dose group (myocardial infarction)., Interpretation: The dose-escalation dosing strategy represents an alternative approach for optimising regorafenib dosing with comparable activity and lower incidence of adverse events and could be implemented in clinical practice on the basis of these data., Funding: Bayer HealthCare Pharmaceuticals., (Copyright © 2019 Elsevier Ltd. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
37. Anal Carcinoma, Version 2.2018, NCCN Clinical Practice Guidelines in Oncology.
- Author
-
Benson AB, Venook AP, Al-Hawary MM, Cederquist L, Chen YJ, Ciombor KK, Cohen S, Cooper HS, Deming D, Engstrom PF, Grem JL, Grothey A, Hochster HS, Hoffe S, Hunt S, Kamel A, Kirilcuk N, Krishnamurthi S, Messersmith WA, Meyerhardt J, Mulcahy MF, Murphy JD, Nurkin S, Saltz L, Sharma S, Shibata D, Skibber JM, Sofocleous CT, Stoffel EM, Stotsky-Himelfarb E, Willett CG, Wuthrick E, Gregory KM, and Freedman-Cass DA
- Subjects
- Anal Canal pathology, Anal Canal surgery, Antineoplastic Combined Chemotherapy Protocols standards, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Anus Neoplasms diagnosis, Anus Neoplasms epidemiology, Anus Neoplasms pathology, Biopsy, Carcinoma, Squamous Cell diagnosis, Carcinoma, Squamous Cell epidemiology, Carcinoma, Squamous Cell pathology, Chemoradiotherapy methods, Chemoradiotherapy standards, Colostomy standards, Disease-Free Survival, Humans, Neoplasm Recurrence, Local diagnosis, Neoplasm Recurrence, Local epidemiology, Neoplasm Recurrence, Local pathology, Patient Care Team standards, Randomized Controlled Trials as Topic, United States epidemiology, Anus Neoplasms therapy, Carcinoma, Squamous Cell therapy, Medical Oncology standards, Neoplasm Recurrence, Local therapy, Societies, Medical standards
- Abstract
The NCCN Guidelines for Anal Carcinoma provide recommendations for the management of patients with squamous cell carcinoma of the anal canal or perianal region. Primary treatment of anal cancer usually includes chemoradiation, although certain lesions can be treated with margin-negative local excision alone. Disease surveillance is recommended for all patients with anal carcinoma because additional curative-intent treatment is possible. A multidisciplinary approach including physicians from gastroenterology, medical oncology, surgical oncology, radiation oncology, and radiology is essential for optimal patient care., (Copyright © 2018 by the National Comprehensive Cancer Network.)
- Published
- 2018
- Full Text
- View/download PDF
38. Rectal Cancer, Version 2.2018, NCCN Clinical Practice Guidelines in Oncology.
- Author
-
Benson AB, Venook AP, Al-Hawary MM, Cederquist L, Chen YJ, Ciombor KK, Cohen S, Cooper HS, Deming D, Engstrom PF, Grem JL, Grothey A, Hochster HS, Hoffe S, Hunt S, Kamel A, Kirilcuk N, Krishnamurthi S, Messersmith WA, Meyerhardt J, Mulcahy MF, Murphy JD, Nurkin S, Saltz L, Sharma S, Shibata D, Skibber JM, Sofocleous CT, Stoffel EM, Stotsky-Himelfarb E, Willett CG, Wuthrick E, Gregory KM, Gurski L, and Freedman-Cass DA
- Subjects
- Antineoplastic Combined Chemotherapy Protocols standards, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Biopsy, Chemoradiotherapy methods, Chemoradiotherapy standards, Disease-Free Survival, Humans, Incidence, Induction Chemotherapy methods, Neoadjuvant Therapy methods, Neoadjuvant Therapy standards, Neoplasm Recurrence, Local diagnosis, Neoplasm Recurrence, Local epidemiology, Neoplasm Recurrence, Local pathology, Neoplasm Staging, Patient Selection, Proctectomy methods, Proctectomy standards, Randomized Controlled Trials as Topic, Rectal Neoplasms diagnosis, Rectal Neoplasms epidemiology, Rectal Neoplasms pathology, Rectum pathology, Rectum surgery, United States epidemiology, Watchful Waiting methods, Watchful Waiting standards, Medical Oncology standards, Neoplasm Recurrence, Local therapy, Rectal Neoplasms therapy, Societies, Medical standards
- Abstract
The NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines) for Rectal Cancer address diagnosis, staging, surgical management, perioperative treatment, management of recurrent and metastatic disease, disease surveillance, and survivorship in patients with rectal cancer. This portion of the guidelines focuses on the management of localized disease, which involves careful patient selection for curative-intent treatment options that sequence multimodality therapy usually comprised of chemotherapy, radiation, and surgical resection., (Copyright © 2018 by the National Comprehensive Cancer Network.)
- Published
- 2018
- Full Text
- View/download PDF
39. Promising New Agents for Colorectal Cancer.
- Author
-
Das S, Ciombor KK, Haraldsdottir S, and Goldberg RM
- Subjects
- Antineoplastic Agents pharmacology, Biomarkers, Tumor, Clinical Trials as Topic, Colorectal Neoplasms etiology, Colorectal Neoplasms pathology, Combined Modality Therapy, Disease Susceptibility, Drug Development, Humans, Microsatellite Instability, Molecular Targeted Therapy, Mutation, Neoplasm Metastasis, Neoplasm Staging, Antineoplastic Agents therapeutic use, Colorectal Neoplasms therapy
- Abstract
Opinion Statement: Choosing the optimal treatment approach for patients with metastatic colorectal cancer (mCRC) demands that oncologists assess both clinical and genomic variables and individualize care based upon the findings. Clinically, choices depend on assessing the side of the colon in which the primary tumor originates, the sites and burden of metastatic disease, the patient's performance status, and their individual comorbidities. Genomic assessment of the tumor to discern the mutational status of genes such as RAS/RAF, HER2, and TRK, as well as assessing whether tumors have defective mismatch repair (dMMR) or high microsatellite instability (MSI-H), all factor in to potential treatment options and can determine clinical trial eligibility. Metastasectomy may be an option for patients with a low burden of disease and accessible liver- or lung-limited metastases. In some unresectable cases, systemic therapy with a FOLFOX- or FOLFIRI-based regimen with or without a biologic agent can lead to sufficient disease reduction to make a patient eligible for resection of metastatic disease. Tumor sidedness and RAS mutational status guide which biologic we add to the initial chemotherapy backbone, with patients with left-sided, RAS wild-type (WT) tumors receiving anti-epidermal growth factor receptor (EGFR)-directed therapy and patients with right-sided tumors or those with RAS mutations receiving bevacizumab. In patients with tumors that manifest microsatellite instability or deficient mismatch repair, we typically administer checkpoint inhibitors such as pembrolizumab or nivolumab after progression on irinotecan- or oxaliplatin-based therapies. In patients with progressive disease, we routinely send tumor tissue for next generation sequencing (NGS) to assess for the presence of actionable genomic alterations such as HER2, BRAF, and TRK fusions and offer them the option of enrollment on clinical trials with agents targeting those or other identified alterations.
- Published
- 2018
- Full Text
- View/download PDF
40. Evaluation of hepatic impairment on pharmacokinetics and safety of crizotinib in patients with advanced cancer.
- Author
-
El-Khoueiry AB, Sarantopoulos J, O'Bryant CL, Ciombor KK, Xu H, O'Gorman M, Chakrabarti J, Usari T, and El-Rayes BF
- Subjects
- Adult, Aged, Area Under Curve, Case-Control Studies, Female, Follow-Up Studies, Humans, Liver Diseases diagnosis, Male, Middle Aged, Neoplasms pathology, Prognosis, Tissue Distribution, Antineoplastic Agents pharmacokinetics, Antineoplastic Agents pharmacology, Crizotinib pharmacokinetics, Crizotinib pharmacology, Liver Diseases metabolism, Neoplasms drug therapy, Severity of Illness Index
- Abstract
Purpose: This phase 1 study evaluated the effect of hepatic impairment on pharmacokinetics and safety of crizotinib in patients with advanced cancer., Methods: Patients were dosed according to hepatic function classified by modified National Cancer Institute Organ Dysfunction Working Group criteria and group assignment [normal (A1 and A2), mild (B), moderate (C1 and C2), or severe (D)]. Primary pharmacokinetic endpoints included area under the concentration-time curve as daily exposure (AUC
daily ) and maximum plasma concentration (Cmax ) at steady state. Safety endpoints included types, incidence, seriousness, and relationship to crizotinib of adverse events., Results: The AUCdaily and Cmax in patients with normal liver function were 7107 ng h/mL and 375.1 ng/mL (A1) and 5422 ng h/mL and 283.9 ng/mL (A2), respectively. The AUCdaily and Cmax ratios of adjusted geometric means for Groups B, C2, and D versus Group A1 were 91.12 and 91.20, 114.08 and 108.87, and 64.47 and 72.63, respectively. Any grade treatment-related adverse events (TRAEs) occurred in 75% of patients; grade 3/4 TRAEs occurred in 25%, including fatigue (6%), hyponatremia (5%), and hyperbilirubinemia (3%)., Conclusions: No adjustment to the approved 250 mg twice daily (BID) dose of crizotinib is recommended for patients with mild hepatic impairment. The recommended dose is 200 mg BID for patients with moderate hepatic impairment, and the dose should not exceed 250 mg daily for patients with severe hepatic impairment. Adverse events appeared consistent among the hepatic impairment groups., Clinical Trial Registration No: NCT01576406.- Published
- 2018
- Full Text
- View/download PDF
41. NCCN Guidelines Insights: Colon Cancer, Version 2.2018.
- Author
-
Benson AB, Venook AP, Al-Hawary MM, Cederquist L, Chen YJ, Ciombor KK, Cohen S, Cooper HS, Deming D, Engstrom PF, Garrido-Laguna I, Grem JL, Grothey A, Hochster HS, Hoffe S, Hunt S, Kamel A, Kirilcuk N, Krishnamurthi S, Messersmith WA, Meyerhardt J, Miller ED, Mulcahy MF, Murphy JD, Nurkin S, Saltz L, Sharma S, Shibata D, Skibber JM, Sofocleous CT, Stoffel EM, Stotsky-Himelfarb E, Willett CG, Wuthrick E, Gregory KM, and Freedman-Cass DA
- Subjects
- Colonic Neoplasms etiology, Humans, Colonic Neoplasms diagnosis, Colonic Neoplasms therapy
- Abstract
The NCCN Guidelines for Colon Cancer provide recommendations regarding diagnosis, pathologic staging, surgical management, perioperative treatment, surveillance, management of recurrent and metastatic disease, and survivorship. These NCCN Guidelines Insights summarize the NCCN Colon Cancer Panel discussions for the 2018 update of the guidelines regarding risk stratification and adjuvant treatment for patients with stage III colon cancer, and treatment of BRAF V600E mutation-positive metastatic colorectal cancer with regimens containing vemurafenib., (Copyright © 2018 by the National Comprehensive Cancer Network.)
- Published
- 2018
- Full Text
- View/download PDF
42. Hypermutated Tumors and Immune Checkpoint Inhibition.
- Author
-
Ciombor KK and Goldberg RM
- Subjects
- Antibodies, Monoclonal, Humanized administration & dosage, Antibodies, Monoclonal, Humanized adverse effects, Antibodies, Monoclonal, Humanized pharmacology, Antibodies, Monoclonal, Humanized therapeutic use, Antineoplastic Agents, Immunological administration & dosage, Antineoplastic Agents, Immunological adverse effects, Antineoplastic Agents, Immunological pharmacology, B7-H1 Antigen antagonists & inhibitors, B7-H1 Antigen immunology, Brain Neoplasms genetics, Brain Neoplasms immunology, Colorectal Neoplasms genetics, Colorectal Neoplasms immunology, Humans, Immunoglobulin G immunology, Immunotherapy, Microsatellite Instability, Mutation, Neoplastic Syndromes, Hereditary genetics, Neoplastic Syndromes, Hereditary immunology, Nivolumab administration & dosage, Nivolumab adverse effects, Nivolumab pharmacology, Nivolumab therapeutic use, Programmed Cell Death 1 Receptor antagonists & inhibitors, Programmed Cell Death 1 Receptor immunology, Antineoplastic Agents, Immunological therapeutic use, Brain Neoplasms therapy, Colorectal Neoplasms therapy, Neoplastic Syndromes, Hereditary therapy
- Abstract
Microsatellite instability-high/DNA mismatch repair deficient tumors are found across the cancer spectrum and often harbor markedly increased numbers of mutations when compared to microsatellite stable/DNA mismatch repair proficient tumors. As a result of this high mutational load, tumor-infiltrating lymphocyte density is increased and more immunogenic neoepitopes are expressed, leading to upregulation of immune checkpoints in these tumors. Checkpoint inhibitors such as pembrolizumab and nivolumab, both immunoglobulin G4 (IgG4) monoclonal antibodies that block interactions between the programmed cell death receptor-1 and its ligands, have significant activity in this tumor class. This review will focus on hypermutated tumors and immuno-oncology drug development for this biologically unique tumor type, with an emphasis on FDA-approved immunotherapies for these cancers, as well as a short discussion of the many therapeutic and scientific challenges ahead in order to optimize the uses of this new class of drug.
- Published
- 2018
- Full Text
- View/download PDF
43. A Comprehensive Review of Sequencing and Combination Strategies of Targeted Agents in Metastatic Colorectal Cancer.
- Author
-
Ciombor KK and Bekaii-Saab T
- Subjects
- Colorectal Neoplasms genetics, Colorectal Neoplasms pathology, Humans, Liver Neoplasms genetics, Liver Neoplasms secondary, Molecular Targeted Therapy, Prognosis, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Biomarkers, Tumor antagonists & inhibitors, Biomarkers, Tumor genetics, Colorectal Neoplasms drug therapy, High-Throughput Nucleotide Sequencing methods, Liver Neoplasms drug therapy, Mutation
- Abstract
The emergence of targeted therapies for the treatment of metastatic colorectal cancer (mCRC) has considerably improved survival, but has also resulted in a dilemma of identifying the optimal sequence and combination of various agents in the mCRC treatment landscape. A number of cytotoxic agents, including irinotecan, oxaliplatin, 5-fluorouracil, capecitabine, and TAS-102, are available for treatment of mCRC. Additionally, whereas patients harboring rat sarcoma viral oncogene homolog ( RAS )-wild type mCRC can be treated with the anti-epidermal growth factor receptor antibodies cetuximab and panitumumab or antiangiogenic agents (bevacizumab, ziv-aflibercept, and ramucirumab), patients with RAS -mutant mCRC are limited to antiangiogenic agents as biologic options. Regorafenib, a multikinase inhibitor, can be used in both RAS subgroups. As such, the recommended sequence of therapies that should be received by each subgroup must also be considered separately. This review provides an overview of recent clinical data for approved and investigational targeted therapies that have been studied across different mCRC treatment lines and patient subgroups. It also examines emerging trends in the treatment landscape for mCRC, including treatment with immune checkpoint inhibitors and the utilization of genomic profiling., Implications for Practice: Currently, there are no established guidelines for optimal sequencing of cytotoxic or targeted agents in metastatic colorectal cancer (mCRC). This review provides a snapshot of the current mCRC treatment paradigm and examines the latest clinical data that support the utilization of several targeted agents alone or in combination with backbone chemotherapy across different lines of treatment and patient populations, highlighting recommendations for their usage. Recent advances in the treatment landscape are also summarized, including genomic profiling and preliminary results with immune checkpoint inhibitors., Competing Interests: Disclosures of potential conflicts of interest may be found at the end of this article., (© AlphaMed Press 2017.)
- Published
- 2018
- Full Text
- View/download PDF
44. Mismatch repair deficiency predicts response of solid tumors to PD-1 blockade.
- Author
-
Le DT, Durham JN, Smith KN, Wang H, Bartlett BR, Aulakh LK, Lu S, Kemberling H, Wilt C, Luber BS, Wong F, Azad NS, Rucki AA, Laheru D, Donehower R, Zaheer A, Fisher GA, Crocenzi TS, Lee JJ, Greten TF, Duffy AG, Ciombor KK, Eyring AD, Lam BH, Joe A, Kang SP, Holdhoff M, Danilova L, Cope L, Meyer C, Zhou S, Goldberg RM, Armstrong DK, Bever KM, Fader AN, Taube J, Housseau F, Spetzler D, Xiao N, Pardoll DM, Papadopoulos N, Kinzler KW, Eshleman JR, Vogelstein B, Anders RA, and Diaz LA Jr
- Subjects
- Adult, Aged, Aged, 80 and over, Antigens, Neoplasm immunology, Brain Neoplasms genetics, Brain Neoplasms mortality, Cell Cycle Checkpoints drug effects, Colorectal Neoplasms genetics, Colorectal Neoplasms mortality, DNA Mismatch Repair, Disease-Free Survival, Female, Humans, Male, Middle Aged, Mutation, Neoplastic Syndromes, Hereditary genetics, Neoplastic Syndromes, Hereditary mortality, Programmed Cell Death 1 Receptor immunology, T-Lymphocytes immunology, Young Adult, Antibodies, Monoclonal, Humanized therapeutic use, Biomarkers, Tumor antagonists & inhibitors, Brain Neoplasms immunology, Brain Neoplasms therapy, Colorectal Neoplasms immunology, Colorectal Neoplasms therapy, Neoplastic Syndromes, Hereditary immunology, Neoplastic Syndromes, Hereditary therapy, Programmed Cell Death 1 Receptor antagonists & inhibitors
- Abstract
The genomes of cancers deficient in mismatch repair contain exceptionally high numbers of somatic mutations. In a proof-of-concept study, we previously showed that colorectal cancers with mismatch repair deficiency were sensitive to immune checkpoint blockade with antibodies to programmed death receptor-1 (PD-1). We have now expanded this study to evaluate the efficacy of PD-1 blockade in patients with advanced mismatch repair-deficient cancers across 12 different tumor types. Objective radiographic responses were observed in 53% of patients, and complete responses were achieved in 21% of patients. Responses were durable, with median progression-free survival and overall survival still not reached. Functional analysis in a responding patient demonstrated rapid in vivo expansion of neoantigen-specific T cell clones that were reactive to mutant neopeptides found in the tumor. These data support the hypothesis that the large proportion of mutant neoantigens in mismatch repair-deficient cancers make them sensitive to immune checkpoint blockade, regardless of the cancers' tissue of origin., (Copyright © 2017 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works.)
- Published
- 2017
- Full Text
- View/download PDF
45. A modified regimen of biweekly gemcitabine and nab-paclitaxel in patients with metastatic pancreatic cancer is both tolerable and effective: a retrospective analysis.
- Author
-
Ahn DH, Krishna K, Blazer M, Reardon J, Wei L, Wu C, Ciombor KK, Noonan AM, Mikhail S, and Bekaii-Saab T
- Abstract
Background: Treatment with nab-paclitaxel with gemcitabine demonstrates a survival advantage when compared with single-agent gemcitabine. However, the combination is associated with significant toxicities, leading to a high rate of drug discontinuation. We implemented a modified regimen of gemcitabine and nab-paclitaxel (mGNabP) in an attempt to minimize toxicities while maintaining efficacy., Methods: A total of 79 evaluable patients with metastatic pancreatic adenocarcinoma (mPC) treated with a modified regimen of gemcitabine (1000 mg/m
2 ) and nab-paclitaxel (125 mg/m2 ) on days 1, 15 of every 28-day cycle were identified from our prospective database. A total of 57 patients received this regimen as first-line treatment and were evaluated for toxicities, progression-free survival (PFS), and overall survival (OS). Overall, 22 patients with advanced or metastatic PC treated with the modified regimen outside the first-line setting were only evaluated for toxicities., Results: The median OS and PFS were 10 months [95% confidence interval (CI) 5.9-13 months] and 5.4 months (95% CI 4.1-7.4 months) for patients that received the modified regimen as first-line therapy. Neurotoxicity occurred in 27% with only 1.6% of patients experiencing grade ⩾3 toxicity. The incidence of grade ⩾3 neutropenia was 19%, resulting in growth factor support in 12% of patients. This rate was similar in patients who received the modified regimen for first-line treatment of mPC versus the overall group., Conclusions: A modified regimen of biweekly nab-paclitaxel with gemcitabine is associated with a lower cost, acceptable toxicity profile and appears to be relatively effective in pancreatic cancer. Prospective randomized studies confirming its potential benefits compared with standard weekly mGNabP are warranted., Competing Interests: Conflict of interest statement: The author(s) declared no potential conflicts of interest with respect to the research, authorship, and/or publication of this article.- Published
- 2017
- Full Text
- View/download PDF
46. Primary Tumor Sidedness as Prognostic and Predictive Biomarker in Metastatic Colorectal Cancer: Further Validation of a Potentially Practice-Changing Variable.
- Author
-
Ciombor KK and Goldberg RM
- Published
- 2017
- Full Text
- View/download PDF
47. A phase I trial investigating pulsatile erlotinib in combination with gemcitabine and oxaliplatin in advanced biliary tract cancers.
- Author
-
Goff LW, Cardin DB, Whisenant JG, Du L, Koyama T, Dahlman KB, Salaria SN, Young RT, Ciombor KK, Gilbert J, Smith SJ, Chan E, and Berlin J
- Subjects
- Adult, Aged, Aged, 80 and over, Antigens, CD, Antineoplastic Agents adverse effects, Antineoplastic Agents therapeutic use, Antineoplastic Combined Chemotherapy Protocols adverse effects, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Biliary Tract Neoplasms genetics, Biliary Tract Neoplasms metabolism, Cadherins metabolism, Deoxycytidine administration & dosage, Deoxycytidine adverse effects, Deoxycytidine therapeutic use, Drug Administration Schedule, ErbB Receptors genetics, Erlotinib Hydrochloride adverse effects, Erlotinib Hydrochloride therapeutic use, Female, Humans, Kaplan-Meier Estimate, Male, Middle Aged, Mutation, Organoplatinum Compounds adverse effects, Organoplatinum Compounds therapeutic use, Oxaliplatin, Proto-Oncogene Proteins p21(ras) genetics, Treatment Outcome, Vimentin metabolism, Gemcitabine, Antineoplastic Agents administration & dosage, Antineoplastic Combined Chemotherapy Protocols administration & dosage, Biliary Tract Neoplasms drug therapy, Deoxycytidine analogs & derivatives, Erlotinib Hydrochloride administration & dosage, Organoplatinum Compounds administration & dosage
- Abstract
Advanced biliary tract cancers (ABTC) are among the deadliest malignancies with limited treatment options after progression on standard-of-care chemotherapy, which includes gemcitabine (GEM) and oxaliplatin (OX). The epidermal growth factor receptor inhibitor erlotinib has been explored in ABTC with modest efficacy. Erlotinib given continuously may antagonize the action of chemotherapy against cycling tumor cells, but pulsatile dosing of erlotinib with chemotherapy may improve efficacy. The purpose of this study was to assess the safety of pulsatile erlotinib with GEMOX. This was a single-institution phase Ib study that enrolled adult patients with unresectable or metastatic biliary tract, pancreas, duodenal, or ampullary carcinomas that have not received any prior treatment for their disease. Dose escalation followed a standard 3 + 3 design, and dose-limiting toxicities (DLTs) were any treatment-related, first course non-hematologic grade ≥ 3 toxicity, except nausea/vomiting, or grade 4 hematologic toxicity. A dose expansion cohort in ABTC was treated at the MTD. Twenty-eight patients were enrolled and 4 dose levels were explored. The MTD was erlotinib 150 mg + GEM 800 mg/m
2 + OX 85 mg/m2 . DLTs were diarrhea and anemia. Most frequent toxicities were nausea (78 %), fatigue (71 %), neuropathy (68 %), and diarrhea (61 %), predominantly grade 1-2. In the ABTC patients, the objective response and disease control rates were 29 % and 94 %, respectively, and median overall survival was 18 months. Erlotinib plus GEMOX was well tolerated. Encouraging anti-tumor activity was seen as evidenced by a high disease control rate and longer median OS than standard chemotherapy in the patients with ABTC., Competing Interests: Compliance with ethical standards Conflicts of interest LWG has served as a consultant for Celgene and has institutional research funding from Astellas Pharma, Pfizer, Onxy, SunPharma, Lilly, and Bristol-Myers Squibb. DBC has served as a consultant for Merrimack and has institutional research funding from Synta, Incyte, Celgene, Hoffman-LaRoxhe, EMD-Serono, and Oncolytics Biotech. KBD has an immediate family member who is employed by Ardent Health Services. KKC has institutional research funding from Pfizer, Boston Biomedical, MedImmune, Onyx, Bayer, Boehringer Ingelheim, and Bristol-Myers Squibb. JG has institutional research funding from AstraZeneca. EC has served on advisory boards for Castle Biosciences, Taiho, EMD-Serono, Amgen, Lilly, Advaxis, Bayer and Merrimack. JB has served as a consultant for Celgene, Genentech, Aduro, Boston Biomedical, Janssen, Cornerstone, Symphogen, and Bayer and has institutional research funding from Genentech, Abbvie, Taiho, Bayer, 5Prime, Phoenix, Incyte, and Vertex. Funding This study was supported in part by Astellas Pharmaceutics, Inc. and the Vanderbilt-Ingram Cancer Center Support Grant (2P30 CA068485–14). Erlotinib was supplied for the study by Astellas Pharmaceutics, Inc. The ITR is supported by the Vanderbilt-Ingram Cancer Center, the TJ Martell Foundation, and the Robert J. Kleberg, Jr. and Helen C. Kleberg Foundation. Ethical approval All procedures performed in studies involving human participants were in accordance with the ethical standards of the institutional and/or national research committee and with the 1964 Helsinki declaration and its later amendments or comparable ethical standards. Informed consent Informed consent was obtained for all individual participants included in the study.- Published
- 2017
- Full Text
- View/download PDF
48. Diagnosis and Diagnostic Imaging of Anal Canal Cancer.
- Author
-
Ciombor KK, Ernst RD, and Brown G
- Subjects
- Anal Canal pathology, Anus Neoplasms pathology, Carcinoma, Squamous Cell, Diagnostic Imaging, Humans, Neoplasm Staging, Reproducibility of Results, Sensitivity and Specificity, Anal Canal diagnostic imaging, Anus Neoplasms diagnostic imaging, Endosonography instrumentation
- Abstract
Anal canal cancer is an uncommon malignancy but one that is often curable with optimal therapy. Owing to its unique location, histology, risk factors, and usual presentation, a careful diagnostic approach is warranted. This approach includes an excellent history and physical examination, including digital rectal examination, laboratory data, and comprehensive imaging. Anal cancer staging and formulation of a treatment plan depends on accurate imaging data. Modern radiographic techniques have improved staging quality and accuracy, and a thorough knowledge of anal anatomy is paramount to the optimal multidisciplinary treatment of this disease., (Copyright © 2016 Elsevier Inc. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
49. Highlights in Gastrointestinal (Colorectal) Cancer Treatment: The Primary Tumor Sidedness Debate and Advances in Immunotherapy.
- Author
-
Ciombor KK and Goldberg RM
- Subjects
- Antibodies, Monoclonal therapeutic use, Antibodies, Monoclonal, Humanized, Colorectal Neoplasms genetics, Colorectal Neoplasms pathology, Disease-Free Survival, Gastrointestinal Neoplasms genetics, Gastrointestinal Neoplasms pathology, Gene Expression Regulation, Neoplastic drug effects, Humans, Neoplasm Proteins biosynthesis, Neoplasm Proteins genetics, Colorectal Neoplasms drug therapy, Gastrointestinal Neoplasms drug therapy, Immunotherapy
- Published
- 2016
- Full Text
- View/download PDF
50. Evaluation of frozen tissue-derived prognostic gene expression signatures in FFPE colorectal cancer samples.
- Author
-
Zhu J, Deane NG, Lewis KB, Padmanabhan C, Washington MK, Ciombor KK, Timmers C, Goldberg RM, Beauchamp RD, and Chen X
- Subjects
- Colorectal Neoplasms pathology, Disease-Free Survival, Female, Formaldehyde, Humans, Male, Neoplasm Recurrence, Local pathology, Oligonucleotide Array Sequence Analysis methods, Paraffin Embedding, Prognosis, Tissue Fixation, Colorectal Neoplasms genetics, Gene Expression Regulation, Neoplastic genetics, Neoplasm Recurrence, Local genetics, Transcriptome genetics
- Abstract
Defining molecular features that can predict the recurrence of colorectal cancer (CRC) for stage II-III patients remains challenging in cancer research. Most available clinical samples are Formalin-Fixed, Paraffin-Embedded (FFPE). NanoString nCounter® and Affymetrix GeneChip® Human Transcriptome Array 2.0 (HTA) are the two platforms marketed for high-throughput gene expression profiling for FFPE samples. In this study, to evaluate the gene expression of frozen tissue-derived prognostic signatures in FFPE CRC samples, we evaluated the expression of 516 genes from published frozen tissue-derived prognostic signatures in 42 FFPE CRC samples measured by both platforms. Based on HTA platform-derived data, we identified both gene (99 individual genes, FDR < 0.05) and gene set (four of the six reported multi-gene signatures with sufficient information for evaluation, P < 0.05) expression differences associated with survival outcomes. Using nCounter platform-derived data, one of the six multi-gene signatures (P < 0.05) but no individual gene was associated with survival outcomes. Our study indicated that sufficiently high quality RNA could be obtained from FFPE tumor tissues to detect frozen tissue-derived prognostic gene expression signatures for CRC patients.
- Published
- 2016
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.