835 results on '"Razelle, Kurzrock"'
Search Results
2. Phase I/II sequencing study of azacitidine, epacadostat, and pembrolizumab in advanced solid tumors
- Author
-
Jason J. Luke, Marwan Fakih, Charles Schneider, E. Gabriela Chiorean, Johanna Bendell, Rebecca Kristeleit, Razelle Kurzrock, Sarah P. Blagden, Irene Brana, Laura W. Goff, Kevin O’Hayer, Ryan Geschwindt, Michael Smith, Feng Zhou, Aung Naing, Institut Català de la Salut, [Luke JJ] UPMC Hillman Cancer Center, Pittsburgh, PA, USA. [Fakih M] City of Hope Comprehensive Cancer Center, Duarte, CA, USA. [Schneider C] Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA. [Chiorean EG] University of Washington School of Medicine, Fred Hutchinson Cancer Research Center, Seattle, WA, USA. [Bendell J] Sarah Cannon Research Institute/Tennessee Oncology, Nashville, TN, USA. [Kristeleit R] Guy’s and St. Thomas’ NHS Foundation Trust, London, UK. [Kurzrock R] University of California San Diego School of Medicine, La Jolla, CA, USA. [Blagden SP] Early Phase Clinical Trials Unit, University of Oxford, Oxford, England, UK. [Brana I] Vall d’Hebron Hospital Universitari, Barcelona, Spain. Vall d’Hebron Institute of Oncology (VHIO), Barcelona, Spain, and Vall d'Hebron Barcelona Hospital Campus
- Subjects
Neoplasms [DISEASES] ,neoplasias [ENFERMEDADES] ,Cancer Research ,Oncology ,Otros calificadores::/uso terapéutico [Otros calificadores] ,Càncer - Tractament ,Medicaments antineoplàstics - Ús terapèutic ,Otros calificadores::Otros calificadores::/farmacoterapia [Otros calificadores] ,acciones y usos químicos::acciones farmacológicas::usos terapéuticos::antineoplásicos [COMPUESTOS QUÍMICOS Y DROGAS] ,Other subheadings::/therapeutic use [Other subheadings] ,Chemical Actions and Uses::Pharmacologic Actions::Therapeutic Uses::Antineoplastic Agents [CHEMICALS AND DRUGS] ,Other subheadings::Other subheadings::/drug therapy [Other subheadings] ,Càncer - Aspectes genètics - Abstract
Background Indoleamine 2,3-dioxygenase 1 (IDO1), an interferon-inducible enzyme, contributes to tumor immune intolerance. Immune checkpoint inhibition may increase interferon levels; combining IDO1 inhibition with immune checkpoint blockade represents an attractive strategy. Epigenetic agents trigger interferon responses and may serve as an immunotherapy priming method. We evaluated whether epigenetic therapy plus IDO1 inhibition and immune checkpoint blockade confers clinical benefit to patients with advanced solid tumors. Methods ECHO-206 was a Phase I/II study where treatment-experienced patients with advanced solid tumors (N = 70) received azacitidine plus an immunotherapy doublet (epacadostat [IDO1 inhibitor] and pembrolizumab). Sequencing of treatment was also assessed. Primary endpoints were safety/tolerability (Phase I), maximum tolerated dose (MTD) or pharmacologically active dose (PAD; Phase I), and investigator-assessed objective response rate (ORR; Phase II). Results In Phase I, no dose-limiting toxicities were reported, the MTD was not reached; a PAD was not determined. ORR was 5.7%, with four partial responses. The most common treatment-related adverse events (AEs) were fatigue (42.9%) and nausea (42.9%). Twelve (17.1%) patients experienced ≥1 fatal AE, one of which (asthenia) was treatment-related. Conclusions Although the azacitidine-epacadostat-pembrolizumab regimen was well tolerated, it was not associated with substantial clinical response in patients with advanced solid tumors previously exposed to immunotherapy.
- Published
- 2023
3. Autoimmune HLA Alleles and Neoepitope Presentation Predict Post-Allogenic Transplant Relapse
- Author
-
Andrea Castro, Aaron M. Goodman, Zachary Rane, James V. Talwar, Garrett M. Frampton, Gerald P. Morris, Scott M. Lippman, Xinlian Zhang, Razelle Kurzrock, and Hannah Carter
- Subjects
Cancer Research ,Oncology ,Immunology ,Immunology and Allergy - Abstract
Introduction Allogeneic hematopoietic stem cell transplantation (allo-HSCT) can cure patients with high-risk myelodysplastic syndromes (MDS) and acute myeloid leukemia (AML). However, many patients relapse or develop debilitating graft-versus-host disease. Transplant restores T-cell reactivity against tumor cells, implicating patient human leukocyte antigen (HLA)-dependent antigen presentation via the major histocompatibility complex as a determinant of response. We sought to identify characteristics of the HLA genotype that influence response in allo-HSCT patients. Methods We collected HLA genotype and panel-based somatic mutation profiles for 55 patients with AML and MDS and available data treated at the University of California San Diego Moores Cancer Center between May 2012 and January 2019. We evaluated characteristics of the HLA genotype relative to relapse-free time and overall survival (OS) post-allo-HSCT using univariable and multivariable regression. Results In multivariable regression, the presence of an autoimmune allele was significantly associated with relapse-free time (hazard ratio [HR], 0.25; p = 0.01) and OS (HR, 0.16; p < 0.005). The better potential of the donor HLA type to present peptides harboring driver mutations trended toward better relapse-free survival (HR, 0.45; p = 0.07) and significantly correlated with longer OS (HR, 0.33; p = 0.01) though only a minority of cases had an HLA mismatch. Conclusion In this single institution retrospective study of patients receiving allo-HSCT for relapsed AML/MDS, characteristics of an individual's HLA genotype (presence of an autoimmune allele and potential of the donor HLA to better present peptides representing driver mutations) were significantly associated with better outcomes. These findings suggest that HLA type may guide the optimal application of allo-HSCT and merit evaluation in larger cohorts. ClinicalTrials.gov Identifier: NCT02478931
- Published
- 2023
- Full Text
- View/download PDF
4. Case Report: Early detection of pancreatic pre-cancer lesion in multimodal approach with exosome liquid biopsy
- Author
-
Harmeet Dhani, Juan Pablo Hinestrosa, Jesus Izaguirre-Carbonell, Heath I. Balcer, Razelle Kurzrock, and Paul R. Billings
- Subjects
Cancer Research ,Oncology - Abstract
BackgroundThe detection of pancreatic ductal adenocarcinoma (PDAC) lesions at pre-cancerous or early-stages is critical to improving patient survival. We have developed a liquid biopsy test (ExoVita®) based on the measurement of protein biomarkers in cancer-derived exosomes. The high sensitivity and specificity of the test for early-stage PDAC has the potential to improve a patient’s diagnostic journey in hopes to impact patient outcomes.MethodsExosome isolation was performed using alternating current electric (ACE) field applied to the patient plasma sample. Following a wash to eliminate unbound particles, the exosomes were eluted from the cartridge. A downstream multiplex immunoassay was performed to measure proteins of interest on the exosomes, and a proprietary algorithm provided a score for probability of PDAC.ResultsWe describe the case of a 60-year-old healthy non-Hispanic white male with acute pancreatitis who underwent numerous invasive diagnostic procedures that failed to detect radiographic evidence of pancreatic lesions. Following the results of our exosome-based liquid biopsy test showing "High Likelihood of PDAC", in addition to KRAS and TP53 mutations, the patient decided to undergo a robotic pancreaticoduodenectomy (Whipple) procedure. Surgical pathology confirmed the diagnosis of high-grade intraductal papillary mucinous neoplasm (IPMN), which was consistent with the results of our ExoVita® test. The patient’s post-operative course was unremarkable. At five-month follow-up, the patient continued to recover well without complications, in addition to a repeat ExoVita test which demonstrated “Low Likelihood of PDAC”.ConclusionThis case report highlights how a novel liquid biopsy diagnostic test based on the detection of exosome protein biomarkers allowed early diagnosis of a high-grade precancerous lesion for PDAC and improved patient outcome.
- Published
- 2023
- Full Text
- View/download PDF
5. <scp>LAG</scp> ‐3 transcriptomic expression patterns across malignancies: Implications for precision immunotherapeutics
- Author
-
Jacob J. Adashek, Shumei Kato, Daisuke Nishizaki, Hirotaka Miyashita, Pradip De, Suzanna Lee, Sarabjot Pabla, Mary Nesline, Jeffrey M. Conroy, Paul DePietro, Scott Lippman, and Razelle Kurzrock
- Subjects
Cancer Research ,Oncology ,Radiology, Nuclear Medicine and imaging - Published
- 2023
- Full Text
- View/download PDF
6. Correlation Between Biomarkers and Treatment Outcomes in Diverse Cancers: A Systematic Review and Meta-Analysis of Phase I and II Immunotherapy Clinical Trials
- Author
-
Elena Fountzilas, Henry Hiep Vo, Peter Mueller, Razelle Kurzrock, and Apostolia-Maria Tsimberidou
- Subjects
Cancer Research ,Oncology - Published
- 2023
- Full Text
- View/download PDF
7. Images in Immunotherapy and Precision Oncology: Angiosarcoma of the Spleen and Liver
- Author
-
Anagha Deshpande, Javier Munoz, Katalin Kelemen, Vrushali Dabak, Amr Hanbali, and Razelle Kurzrock
- Subjects
Cancer Research ,Oncology ,Immunology ,Immunology and Allergy - Abstract
Primary splenic or hepatic angiosarcomas are ultra-rare and aggressive malignancies associated with poor prognosis. The mainstay treatments are surgical resection and chemotherapy. We report a case of angiosarcoma in a 50-year-old woman who presented with bruising, fatigue, ecchymosis, and hepatosplenomegaly. She was treated with the multi-kinase inhibitor sunitinib for 4 weeks before developing a splenic hemorrhage and succumbing. Recent studies have demonstrated the clinical benefit of immunotherapies in angiosarcomas. Additionally, sequencing techniques have showcased the diverse molecular aberrations involved in angiosarcomas, which offer opportunities for precision-matched targeted therapies such as inhibitors of the VEGF/VEGFR axis and PI3K/Akt/mTor pathway.
- Published
- 2022
- Full Text
- View/download PDF
8. Variable Mutation Expression in Human Cancers: A 'Hide-and-Seek' Mechanism Linked to Differential MHC-I Presentation Dynamics
- Author
-
Amélie Boichard and Razelle Kurzrock
- Subjects
Antigen Presentation ,Cancer Research ,Oncology ,Neoplasms ,Histocompatibility Antigens Class I ,Mutation ,Mutation, Missense ,Humans ,Prognosis - Abstract
Not all genomic mutations are expressed at the transcript/protein level, which may explain variation in cancer development, prognosis, and treatment response/resistance. In this study, our aim was to describe the prevalence of somatic mutation loss of expression (‘variant silencing’) in a large collection of human samples, and the potential impact of such variant silencing on tumor immunogenicity. Whole-exome mutation description and tumor-normal paired mRNA expression data originating from 636 unique patients diagnosed with 21 distinct tumor types (all solid tumors) were retrieved from The Cancer Genome Atlas (TCGA). Antigenicity and immunogenicity of neopeptides originating from mutated proteins within a same tumor sample were predicted using the tools available from the Immune Epitope Database (IEDB). A total of 65,072 missense mutations were studied. We demonstrated that 9.06% (N = 10,604 silenced/117,505 total variants) somatic variants were silenced in human tumors. Transciptomic silencing is significantly associated with proteins presenting better peptide processing, MHC-I binding, and T-cell recognition; and is more likely observed in lymphocyte-depleted tumors. Silencing may participate in tumor resistance by clonal selection and immune evasion. In the era of precision medicine, we suggest that therapeutic choices should be informed by both the presence of a genomic mutation and its actual transcript expression.
- Published
- 2022
- Full Text
- View/download PDF
9. ERBB family fusions are recurrent and actionable oncogenic targets across cancer types
- Author
-
Laura Schubert, Andrew Elliott, Anh T. Le, Adriana Estrada-Bernal, Robert C. Doebele, Emil Lou, Hossein Borghaei, Michael J. Demeure, Razelle Kurzrock, Joshua E. Reuss, Sai-Hong Ignatius Ou, David R. Braxton, Christian A. Thomas, Sourat Darabi, Wolfgang Michael Korn, Wafik S. El-Deiry, and Stephen V. Liu
- Subjects
Cancer Research ,Oncology - Abstract
PurposeGene fusions involving receptor tyrosine kinases (RTKs) define an important class of genomic alterations with many successful targeted therapies now approved for ALK, ROS1, RET and NTRK gene fusions. Fusions involving the ERBB family of RTKs have been sporadically reported, but their frequency has not yet been comprehensively analyzed and functional characterization is lacking on many types of ERBB fusions.Materials and methodsWe analyzed tumor samples submitted to Caris Life Sciences (n=64,354), as well as the TCGA (n=10,967), MSK IMPACT (n=10,945) and AACR GENIE (n=96,324) databases for evidence of EGFR, ERBB2 and ERBB4 gene fusions. We also expressed several novel fusions in cancer cell lines and analyzed their response to EGFR and HER2 tyrosine kinase inhibitors (TKIs).ResultsIn total, we identified 1,251 ERBB family fusions, representing an incidence of approximately 0.7% across all cancer types. EGFR, ERBB2, and ERBB4 fusions were most frequently found in glioblastoma, breast cancer and ovarian cancer, respectively. We modeled two novel types of EGFR and ERBB2 fusions, one with a tethered kinase domain and the other with a tethered adapter protein. Specifically, we expressed EGFR-ERBB4, EGFR-SHC1, ERBB2-GRB7 and ERBB2-SHC1, in cancer cell lines and demonstrated that they are oncogenic, regulate downstream signaling and are sensitive to small molecule inhibition with EGFR and HER2 TKIs.ConclusionsWe found that ERBB fusions are recurrent mutations that occur across multiple cancer types. We also establish that adapter-tethered and kinase-tethered fusions are oncogenic and can be inhibited with EGFR or HER2 inhibitors. We further propose a nomenclature system to categorize these fusions into several functional classes.
- Published
- 2023
- Full Text
- View/download PDF
10. Concordance between cancer gene alterations in tumor and circulating tumor <scp>DNA</scp> correlates with poor survival in a real‐world precision‐medicine population
- Author
-
Shai Rosenberg, Gil Ben Cohen, Shumei Kato, Ryosuke Okamura, Scott M. Lippman, and Razelle Kurzrock
- Subjects
Cancer Research ,Oncology ,Genetics ,Molecular Medicine ,General Medicine - Published
- 2023
- Full Text
- View/download PDF
11. Universal Germline and Tumor Genomic Testing Needed to Win the War Against Cancer: Genomics Is the Diagnosis
- Author
-
Razelle Kurzrock and Vivek Subbiah
- Subjects
Cancer Research ,Oncology - Published
- 2023
- Full Text
- View/download PDF
12. Of Mice, Not Men: When the Bench-to-Bedside Bridge Is Broken
- Author
-
Mina, Nikanjam, Shumei, Kato, and Razelle, Kurzrock
- Subjects
Cancer Research ,Oncology ,Immunology ,Immunology and Allergy - Published
- 2022
- Full Text
- View/download PDF
13. Establishment of Patient-Derived Succinate Dehydrogenase–Deficient Gastrointestinal Stromal Tumor Models for Predicting Therapeutic Response
- Author
-
Sunil J. Advani, Hyunho Yoon, Rowan Ustoy, Mayra Yebra, Vipul R. Sheth, Sudeep Banerjee, Michael Heinrich, Christian M. Metallo, Joseph A. Aguilera, Sangkyu Noh, Scott M. Lippman, Razelle Kurzrock, Chih-Min Tang, Olivier Harismendy, Paul T. Fanta, Adam M. Burgoyne, Avi Kumar, Thekla Cordes, Shruti Bhargava, Sam Li, Jason K. Sicklick, and Christopher L. Corless
- Subjects
Cancer Research ,Receptor, Platelet-Derived Growth Factor alpha ,Adolescent ,Gastrointestinal Stromal Tumors ,Oncology and Carcinogenesis ,PDGFRA ,Article ,Young Adult ,03 medical and health sciences ,Rare Diseases ,0302 clinical medicine ,Clinical Research ,Humans ,Medicine ,Oncology & Carcinogenesis ,Stromal tumor ,neoplasms ,Gastrointestinal Neoplasms ,Cancer ,030304 developmental biology ,0303 health sciences ,Gastrointestinal tract ,Temozolomide ,GiST ,business.industry ,Platelet-Derived Growth Factor alpha ,medicine.disease ,digestive system diseases ,3. Good health ,Succinate Dehydrogenase ,Clinical trial ,Proto-Oncogene Proteins c-kit ,Orphan Drug ,Good Health and Well Being ,Oncology ,030220 oncology & carcinogenesis ,Mutation ,Cancer research ,Sarcoma ,Digestive Diseases ,business ,Tyrosine kinase ,Receptor ,medicine.drug - Abstract
Purpose: Gastrointestinal stromal tumor (GIST) is the most common sarcoma of the gastrointestinal tract, with mutant succinate dehydrogenase (SDH) subunits (A–D) comprising less than 7.5% (i.e., 150–200/year) of new cases annually in the United States. Contrary to GISTs harboring KIT or PDGFRA mutations, SDH-mutant GISTs affect adolescents/young adults, often metastasize, and are frequently resistant to tyrosine kinase inhibitors (TKI). Lack of human models for any SDH-mutant tumors, including GIST, has limited molecular characterization and drug discovery. Experimental Design: We describe methods for establishing novel patient-derived SDH-mutant (mSDH) GIST models and interrogated the efficacy of temozolomide on these tumor models in vitro and in clinical trials of patients with mSDH GIST. Results: Molecular and metabolic characterization of our patient-derived mSDH GIST models revealed that these models recapitulate the transcriptional and metabolic hallmarks of parent tumors and SDH deficiency. We further demonstrate that temozolomide elicits DNA damage and apoptosis in our mSDH GIST models. Translating our in vitro discovery to the clinic, a cohort of patients with SDH-mutant GIST treated with temozolomide (n = 5) demonstrated a 40% objective response rate and 100% disease control rate, suggesting that temozolomide represents a promising therapy for this subset of GIST. Conclusions: We report the first methods to establish patient-derived mSDH tumor models, which can be readily employed for understanding patient-specific tumor biology and treatment strategies. We also demonstrate that temozolomide is effective in patients with mSDH GIST who are refractory to existing chemotherapeutic drugs (namely, TKIs) in clinic for GISTs, bringing a promising treatment option for these patients to clinic. See related commentary by Blakely et al., p. 3
- Published
- 2022
- Full Text
- View/download PDF
14. SMARCA4: Implications of an Altered Chromatin-Remodeling Gene for Cancer Development and Therapy
- Author
-
Kristina Mardinian, Jacob J. Adashek, Gregory P. Botta, Shumei Kato, and Razelle Kurzrock
- Subjects
Cancer Research ,Oncology ,SMARCA4 Gene ,EZH2 ,Cancer research ,SMARCA4 ,Synthetic lethality ,Biology ,Article ,Chromatin remodeling ,Germline ,Loss function ,Immune checkpoint - Abstract
The SWI/SNF chromatin remodeling complex, via nucleosome topology modulation, regulates transcription. The SMARCA4 (BRG1) subunit codes for the ATPase energy engine of the SWI/SNF complex. SMARCA4 is a tumor suppressor that is aberrant in ∼5% to 7% of human malignancies. Class I SMARCA4 alterations (truncating mutations, fusions, and homozygous deletion) lead to loss of function whereas class II alterations (missense mutations) have a dominant negative/gain-of-function effect and/or loss-of function. SMARCA4 alterations typify the ultra-rare small cell carcinomas of the ovary hypercalcemic type (SCCOHT) and SMARCA4-deficient thoracic and uterine sarcomas; they are also found in a subset of more common tumors, for example, lung, colon, bladder, and breast carcinomas. Germline variants in the SMARCA4 gene lead to various hereditary conditions: rhabdoid tumor predisposition syndrome-2 (RTPS2), characterized by loss-of-function alterations and aggressive rhabdoid tumors presenting in infants and young children; and Coffin-Siris syndrome, characterized by dominant negative/gain-of function alterations and developmental delays, microcephaly, unique facies, and hypoplastic nails of the fifth fingers or toes. A minority of rhabdoid tumors have a germline SMARCA4 variant as do >40% of women with SCCOHT. Importantly, immune checkpoint blockade has shown remarkable, albeit anecdotal, responses in SCCOHT. In addition, there is ongoing research into BET, EZH2, HDAC, CDK4/6, and FGFR inhibitors, as well as agents that might induce synthetic lethality via DNA damage repair impairment (ATR inhibitors and platinum chemotherapy), or via the exploitation of mitochondrial oxidative phosphorylation inhibitors or AURKA inhibitors, in SMARCA4-aberrant cancers.
- Published
- 2021
- Full Text
- View/download PDF
15. Genome-wide Sequencing of Cell-free DNA Enables Detection of Copy-number Alterations in Patients with Cancer Where Tissue Biopsy is Not Feasible
- Author
-
Graham McLennan, Amin Mazloom, Gregory A. Daniels, Daniel S. Grosu, Marcia Eisenberg, Razelle Kurzrock, Kimberly A. Holden, Shumei Kato, Prachi Nakashe, Kerry D. Fitzgerald, Eyad Almasri, Taylor J. Jensen, Christopher K. Ellison, Erin McCarthy, Lisa Kim, and Aaron M. Goodman
- Subjects
Adult ,Male ,Genome instability ,Oncology ,Cancer Research ,medicine.medical_specialty ,DNA Copy Number Variations ,Genome ,Young Adult ,Neoplasms ,Internal medicine ,Biomarkers, Tumor ,medicine ,Humans ,In patient ,Precision Medicine ,Aged ,Aged, 80 and over ,Alternative methods ,Autosome ,Whole Genome Sequencing ,business.industry ,Cancer ,Middle Aged ,medicine.disease ,Cell-free fetal DNA ,Female ,business ,Cell-Free Nucleic Acids ,Tissue biopsy - Abstract
When tissue biopsy is not medically prudent or tissue is insufficient for molecular testing, alternative methods are needed. Because cell-free DNA (cfDNA) has been shown to provide a representative surrogate for tumor tissue, we sought to evaluate its utility in this clinical scenario. cfDNA was isolated from the plasma of patients and assayed with low-coverage (∼0.3×), genome-wide sequencing. Copy-number alterations (CNA) were identified and characterized using analytic methods originally developed for noninvasive prenatal testing (NIPT) and quantified using the genomic instability number (GIN), a metric that reflects the quantity and magnitude of CNAs across the genome. The technical variability of the GIN was first evaluated in an independent cohort comprising genome-wide sequencing results from 27,754 women who consented to have their samples used for research and whose NIPT results yielded no detected CNAs to establish a detection threshold. Subsequently, cfDNA sequencing data from 96 patients with known cancers but for whom a tissue biopsy could not be obtained are presented. An elevated GIN was detected in 35% of patients and detection rates varied by tumor origin. Collectively, CNAs covered 96.6% of all autosomes. Survival was significantly reduced in patients with an elevated GIN relative to those without. Overall, these data provide a proof of concept for the use of low-coverage, genome-wide sequencing of cfDNA from patients with cancer to obtain relevant molecular information in instances where tissue is difficult to access. These data may ultimately serve as an informative complement to other molecular tests.
- Published
- 2021
- Full Text
- View/download PDF
16. Pan-cancer molecular tumor board experience with biomarker-driven precision immunotherapy
- Author
-
Bryan H. Louie, Shumei Kato, Ki Hwan Kim, Hyo Jeong Lim, Ryosuke Okamura, Ramez N. Eskander, Gregory Botta, Hitendra Patel, Suzanna Lee, Scott M. Lippman, Jason K. Sicklick, and Razelle Kurzrock
- Subjects
Cancer Research ,Oncology - Abstract
Despite remarkable responses to immune checkpoint blockade (ICB) in some advanced cancers, most patients do not benefit, perhaps due to the complexity of tumor/immune/genome interactions. We implemented a multidisciplinary Molecular Tumor Board (MTB) that reviewed multi-omic cancer characteristics to develop N-of-One therapies for patients in the pan-cancer, advanced, refractory setting. This study evaluates the experience of 80 patients who were presented to the MTB and received a treatment regimen that included ICB. Overall, 60/80 patients (75%) who received ICB following MTB discussion had a high degree of matching between tumor molecular characteristics, including ICB biomarkers (reflected by a high Matching Score (≥50%)) and therapy administered. Patients with high versus low Matching Score experienced significantly longer median progression-free survival (6.4 vs. 3.0 months; p = 0.011) and median overall survival (15.3 vs. 4.7 months; p = 0.014) and higher clinical benefit rates (stable disease ≥6 months/partial response/complete response) (53% vs. 21%, p = 0.019). Although most patients (52/80 (65%)) received a personalized combination therapy (e.g., targeted, hormonal, chemotherapy, or a second immunotherapy agent), administering >1 drug was not associated with outcome. Only degree of matching and age, but no other variables, including individual biomarkers (e.g., microsatellite status, tumor mutational burden, or PD-L1 status), were independently correlated with outcome. In the pan-cancer setting, the MTB facilitated a precision medicine strategy to match therapeutic regimens that included ICB alone or combined with matched targeted drugs to patients with advanced malignancy, which was associated with improved clinical outcomes.
- Published
- 2022
- Full Text
- View/download PDF
17. Prevalence of ARID1A Mutations in Cell-Free Circulating Tumor DNA in a Cohort of 71,301 Patients and Association with Driver Co-Alterations
- Author
-
Razelle Kurzrock, Charu Aggarwal, Caroline Weipert, Lesli Kiedrowski, Jonathan Riess, Heinz-Josef Lenz, and David Gandara
- Subjects
Cancer Research ,Oncology ,ARID1A ,cancer ,liquid biopsy ,cfDNA - Abstract
ARID1A abnormalities disturb transcriptional processes regulated by chromatin remodeling and correlate with immunotherapy responsiveness. We report the first blood-based cell-free DNA (cfDNA) next-generation sequencing (NGS) ARID1A analysis. From November 2016 through August 2019, 71,301 patients with advanced solid tumors underwent clinical blood-derived cfDNA testing. Of these patients, 62,851 (88%) had ≥1 cfDNA alteration, and 3137 (of the 62,851) (5%) had ≥1 deleterious ARID1A alteration (a frequency similar to the ~6% generally reported in tissue NGS), suggesting this non-invasive test’s value in interrogating ARID1A. ARID1A cfDNA alterations were most frequent in endometrial cancer, 21.3% of patients; bladder cancer, 12.9%; gastric cancer, 11%; cholangiocarcinoma, 10.9%; and hepatocellular carcinoma, 10.6%. Blood samples with a functional ARID1A abnormality had more alterations/sample (median, 6 versus 4; p < 0.0001) and more frequent co-alterations in ≥1 gene in key oncogenic pathways: signal transduction, RAS/RAF/MAPK, PI3K/Akt/mTor, and the cell cycle. Taken together, our data suggest that liquid (blood) biopsies identify ARID1A alterations at a frequency similar to that found in primary tumor material. Furthermore, co-alterations in key pathways, some of which are pharmacologically tractable, occurred more frequently in samples with functional (deleterious) ARID1A alterations than in those without such aberrations, which may inform therapeutic strategies.
- Published
- 2022
- Full Text
- View/download PDF
18. Osteosarcoma with cell-cycle and fibroblast growth factor genomic alterations: case report of Molecular Tumor Board combination strategy resulting in long-term exceptional response
- Author
-
Hanna E. Persha, Shumei Kato, Pradip De, Jacob J. Adashek, Jason K. Sicklick, Vivek Subbiah, and Razelle Kurzrock
- Subjects
Cancer Research ,Oncology ,Hematology ,Molecular Biology - Abstract
There is a paucity of information about molecularly driven therapy in osteosarcomas. We report a 31-year-old woman with chemotherapy–refractory metastatic osteosarcoma who was successfully treated with the combination of palbociclib (CDK4/6 inhibitor) and lenvatinib (multikinase FGFR inhibitor), selected based on next generation sequencing that showed CDK4 and CCND2 amplifications (upregulates CDK4/6), and FGF6 (ligand for FGFR1,2 and 4), FGF23 (ligand for FGFR1,2,3, and 4) and FRS2 (adaptor protein for FGFR signaling) amplifications. The patient’s tumor showed 68% reduction in positron emission tomography (PET) avidity, lasting 31 months after therapy initiation, when a solitary recurrence occurred, was resected, and treatment continued. The patient remains on matched targeted therapy at 51 + months from the start of the combination. Treatment was given at reduced dosing (lenvatinib 10 mg oral daily (approved dose = 24 mg daily)) and palbociclib 75 mg oral daily, one week on and one week off (approved dose = 125 mg oral daily, three weeks on/one week off) and is tolerated well. Therefore, co-targeting the aberrant cyclin and FGFR pathways resulted in long-term exceptional response in a patient with refractory advanced osteosarcoma.
- Published
- 2022
- Full Text
- View/download PDF
19. Comparative Genomic Analysis of Intrahepatic Cholangiocarcinoma: Biopsy Type, Ancestry, and Testing Patterns
- Author
-
Jason K. Sicklick, Hanna Tukachinsky, Kimberly McGregor, Shumei Kato, Mason A. Israel, Halla Nimeiri, Geoffrey R. Oxnard, Jeffrey S. Ross, Karthikeyan Murugesan, Ethan Sokol, Razelle Kurzrock, Ole Gjoerup, and Natalie Danziger
- Subjects
0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,IDH1 ,Biopsy ,Cholangiocarcinoma ,03 medical and health sciences ,0302 clinical medicine ,Text mining ,Internal medicine ,medicine ,Humans ,Liquid biopsy ,Intrahepatic Cholangiocarcinoma ,medicine.diagnostic_test ,business.industry ,Genomics ,medicine.disease ,Primary tumor ,Clinical trial ,Bile Ducts, Intrahepatic ,030104 developmental biology ,Bile Duct Neoplasms ,030220 oncology & carcinogenesis ,Biomarker (medicine) ,Hepatobiliary ,business - Abstract
BackgroundAt diagnosis, the majority of patients with intrahepatic cholangiocarcinoma (IHCC) present with advanced disease and a poor prognosis. Comprehensive genomic profiling (CGP) early in the disease course may increase access to targeted therapies and clinical trials; however, unresolved issues remain surrounding the optimal biopsy type to submit for CGP.Patients and MethodsMutational frequencies between primary tumor biopsies (Pbx), metastatic biopsies (Mbx), and liquid biopsies (Lbx) in 1,632 patients with IHCC were compared.ResultsPotentially actionable alterations were found in 52%, 34%, and 35% of patients in the Pbx, Mbx, and Lbx cohorts, respectively. In Pbx, Mbx, and Lbx, FGFR2 rearrangements were found in 9%, 6%, and 4%, and IDH1 mutations were identified in 16%, 5%, and 9% patients, respectively. Moreover, alterations in FGFR2 and IDH1 were significantly associated with distinct ancestries, including 2.1-fold enrichment for FGFR2 rearrangements in patients with African ancestry and 1.5-fold enrichment for IDH1 mutations in patients with admixed American (Hispanic) ancestry. Finally, the publication of biomarker-driven clinical trials in IHCC correlated with changing CGP testing patterns. Significant correlations between patient characteristics and IHCC trial disclosures were observed, including a significant decrease from time between biopsy and CGP testing, and more frequent testing of primary versus metastatic samples.ConclusionOverall, because of the high likelihood of identifying actionable genomic alterations, CGP should be considered for the majority of patients with inoperable IHCC, and Lbx and Mbx can be considered as part of the diagnostic suite.Implications for PracticeComprehensive genomic profiling (CGP) should be considered for all patients with intrahepatic cholangiocarcinoma (IHCC) or suspected IHCC, as actionable alterations were commonly found in multiple genes and a wide variety of FGFR2 fusion partners were identified. The disclosure of IHCC trial data correlated with increased use of CGP, an encouraging trend that moves new therapeutic options forward for rare cancers with a rare biomarker. Although tissue from the primary lesion may identify actionable alterations at higher rates, CGP of a liquid biopsy or metastatic site can be considered, particularly if the primary tissue block is exhausted.
- Published
- 2021
- Full Text
- View/download PDF
20. Functional measurement of mitogen-activated protein kinase pathway activation predicts responsiveness of RAS-mutant cancers to MEK inhibitors
- Author
-
Gabi Tarcic, Razelle Kurzrock, Ori Zelichov, Shumei Kato, Suzanna Lee, Michael Vidne, Ryosuke Okamura, and Robert Porter
- Subjects
Adult ,Male ,0301 basic medicine ,MAPK/ERK pathway ,Cancer Research ,Time Factors ,Cell ,Mutant ,Antineoplastic Agents ,03 medical and health sciences ,0302 clinical medicine ,Neoplasms ,medicine ,Humans ,Genetic Predisposition to Disease ,Protein kinase A ,Protein Kinase Inhibitors ,Aged ,Aged, 80 and over ,biology ,business.industry ,MEK inhibitor ,Cancer ,Transfection ,Middle Aged ,MAP Kinase Kinase Kinases ,medicine.disease ,Progression-Free Survival ,Genes, ras ,Phenotype ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,030220 oncology & carcinogenesis ,Mitogen-activated protein kinase ,Mutation ,biology.protein ,Cancer research ,Female ,Mitogen-Activated Protein Kinases ,business ,Signal Transduction - Abstract
Background RAS variant–related functional impact on the mitogen-activated protein kinase (MAPK) pathway, and correlation between MAPK activation and MAPK/ERK kinase (MEK) inhibitor responsiveness, is not established. Patients and methods Of 1,693 tumours sequenced, 576 harboured a RAS alteration; 62 patients received an MEK inhibitor (MEKi) and had RAS mutations that were functionally characterised. We report that RAS mutants have variable levels of MAPK activity, as measured by a functional cell-based assay that quantified MAPK pathway activation after transfection with a variety of RAS mutations. Results Patients with tumours harbouring RAS alterations with high versus low MAPK activity who were treated with an MEKi showed significantly longer median progression-free survival (PFS) (5.0 vs. 2.3 months; p = 0.0034) and overall survival (20.0 vs. 5.0 months; p = 0.0146) and a trend towards higher rates of clinical benefit (stable disease ≥6 months or partial/complete remission) (38% versus 15%; p = 0.095) (p-values as per univariate analysis). PFS remained statistically significant after the multivariate analysis (p = 0.003). Conclusions These results support a correlation between RAS-mutant cancers with greater MAPK signalling and PFS after MEKi treatment.
- Published
- 2021
- Full Text
- View/download PDF
21. KRAS‐Mutated, Estrogen Receptor‐Positive Low‐Grade Serous Ovarian Cancer: Unraveling an Exceptional Response Mystery
- Author
-
Edward C. Stites, Steven C. Plaxe, Thomas McFall, Ramez N. Eskander, Kasey Bamel, Sadakatsu Ikeda, Barbara A. Parker, Shumei Kato, Razelle Kurzrock, and Kenta Takahashi
- Subjects
0301 basic medicine ,Cancer Research ,medicine.drug_class ,Estrogen receptor ,Personalized therapy ,Proto-Oncogene Proteins p21(ras) ,03 medical and health sciences ,Phosphatidylinositol 3-Kinases ,0302 clinical medicine ,Nitriles ,medicine ,KRAS ,Humans ,Trametinib ,Ovarian Neoplasms ,Aromatase inhibitor ,business.industry ,Aromatase Inhibitors ,Letrozole ,MEK inhibitor ,Triazoles ,medicine.disease ,Estrogen ,Tamoxifen ,030104 developmental biology ,Oncology ,Receptors, Estrogen ,030220 oncology & carcinogenesis ,Combination ,Cancer research ,Precision Medicine Clinic: Molecular Tumor Board ,Female ,business ,Ovarian cancer ,medicine.drug - Abstract
We report on a woman with aggressive estrogen receptor‐positive, KRAS‐mutated ovarian cancer who achieved a remarkable response to combination therapy with the MEK inhibitor (trametinib) and the aromatase inhibitor (letrozole), even though the disease had failed to respond to a combination of a PI3K inhibitor and different MEK inhibitor, as well as to trametinib and the estrogen modulator, tamoxifen, and to letrozole by itself. The mechanism of action for exceptional response was elucidated by in vitro experiments that demonstrated that the fact that tamoxifen can have an agonistic effect in addition to antagonist activity, whereas letrozole results only in estrogen depletion was crucial to the response achieved when letrozole was combined with an MEK inhibitor. Our current observations indicate that subtle variations in mechanisms of action of outwardly similar regimens may have a major impact on outcome and that such translational knowledge is critical for optimizing a precision medicine strategy. Key Points This report describes the remarkable response of a patient with KRAS‐mutated, estrogen receptor‐positive low‐grade serous ovarian cancer treated with trametinib (MEK inhibitor) and letrozole (aromatase inhibitor), despite prior progression on similar agents including tamoxifen (estrogen modulator).In vitro investigation revealed that tamoxifen can have agonistic in addition to antagonistic effects, which could be the reason for the patient not responding to the combination of trametinib and tamoxifen.The current observations suggest that drugs with different mechanisms of action targeting the same receptor may have markedly different anticancer activity when used in combinations., This article reports the case of a patient with aggressive estrogen receptor‐positive, KRAS‐mutated ovarian cancer who achieved a remarkable response to combination therapy with the MEK inhibitor trametinib and the aromatase inhibitor letrozole, despite earlier failures of treatment with other combination inhibitor treatment. This article focuses on the possibility that subtle variations in mechanisms of action of outwardly similar regimens may have major effects on patient outcomes.
- Published
- 2021
22. The Challenges of Tumor Mutational Burden as an Immunotherapy Biomarker
- Author
-
Debora de Melo Gagliato, Razelle Kurzrock, Denis Leonardo Fontes Jardim, and Aaron M. Goodman
- Subjects
0301 basic medicine ,Cancer Research ,T-Lymphocytes ,medicine.medical_treatment ,Immune checkpoint inhibitors ,Major histocompatibility complex ,medicine.disease_cause ,Article ,Major Histocompatibility Complex ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Receptor repertoire ,Neoplasms ,Biomarkers, Tumor ,medicine ,Humans ,Immune Checkpoint Inhibitors ,Mutation ,biology ,business.industry ,Cancer ,Immunotherapy ,medicine.disease ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Cancer research ,biology.protein ,Biomarker (medicine) ,business - Abstract
Summary Tumor mutational burden (TMB) reflects cancer mutation quantity. Mutations are processed to neo-antigens and presented by major histocompatibility complex (MHC) proteins to T cells. To evade immune eradication, cancers exploit checkpoints that dampen T cell reactivity. Immune checkpoint inhibitors (ICIs) have transformed cancer treatment by enabling T cell reactivation; however, response biomarkers are required, as most patients do not benefit. Higher TMB results in more neo-antigens, increasing chances for T cell recognition, and clinically correlates with better ICI outcomes. Nevertheless, TMB is an imperfect response biomarker. A composite predictor that also includes critical variables, such as MHC and T cell receptor repertoire, is needed.
- Published
- 2021
- Full Text
- View/download PDF
23. A WIN Consortium phase I study exploring avelumab, palbociclib, and axitinib in advanced non-small cell lung cancer
- Author
-
Benjamin Solomon, Ana Callejo, Jair Bar, Guy Berchem, Lyudmila Bazhenova, Pierre Saintigny, Fanny Wunder, Jacques Raynaud, Nicolas Girard, J. Jack Lee, Raed Sulaiman, Bruce Prouse, Catherine Bresson, Hila Ventura, Shai Magidi, Eitan Rubin, Brandon Young, Amir Onn, Brian Leyland‐Jones, Richard L. Schilsky, Vladimir Lazar, Enriqueta Felip, Razelle Kurzrock, Institut Català de la Salut, [Solomon B] Avera Cancer Institute, Sioux Falls, South Dakota, USA. [Callejo A, Felip E] Vall d'Hebron Hospital Universitari, Barcelona, Spain. Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain. [Bar J] Chaim Sheba Medical Center, Tel Hashomer, Israel. [Berchem G] Centre Hospitalier de Luxembourg, Luxembourg Institute of Health, Luxembourg City, Luxemburg. [Bazhenova L] University of California San Diego, Moores Cancer Center, San Diego, California, USA. [Saintigny P] Centre Léon Bérard, Cancer Research Center of Lyon, University of Lyon, Lyon, France, and Vall d'Hebron Barcelona Hospital Campus
- Subjects
Cancer Research ,Lung Neoplasms ,anti-PD-L1 ,CDK4 ,Axitinib ,Pyridines ,Clinical Trials and Supportive Activities ,Oncology and Carcinogenesis ,Otros calificadores::Otros calificadores::/farmacoterapia [Otros calificadores] ,Other subheadings::Other subheadings::/drug therapy [Other subheadings] ,Antibodies, Monoclonal, Humanized ,NSCLC ,Quimioteràpia combinada ,Piperazines ,Antibodies ,B7-H1 Antigen ,VEGFR ,transcriptomics ,Clinical Research ,Therapeutics::Therapeutics::Drug Therapy::Antineoplastic Protocols::Therapeutics::Drug Therapy::Antineoplastic Combined Chemotherapy Protocols [ANALYTICAL, DIAGNOSTIC AND THERAPEUTIC TECHNIQUES, AND EQUIPMENT] ,Carcinoma, Non-Small-Cell Lung ,Proto-Oncogene Proteins ,Antineoplastic Combined Chemotherapy Protocols ,Monoclonal ,CDK4/6 ,genomics ,Humans ,Radiology, Nuclear Medicine and imaging ,Non-Small-Cell Lung ,Humanized ,Lung ,Cancer ,Carcinoma ,Lung Cancer ,terapéutica::terapéutica::farmacoterapia::protocolos antineoplásicos::terapéutica::farmacoterapia::protocolos de quimioterapia antineoplásica combinada [TÉCNICAS Y EQUIPOS ANALÍTICOS, DIAGNÓSTICOS Y TERAPÉUTICOS] ,Antibodies, Monoclonal ,Evaluation of treatments and therapeutic interventions ,Neoplasms::Neoplasms by Site::Thoracic Neoplasms::Respiratory Tract Neoplasms::Lung Neoplasms::Bronchial Neoplasms::Carcinoma, Bronchogenic::Carcinoma, Non-Small-Cell Lung [DISEASES] ,neoplasias::neoplasias por localización::neoplasias torácicas::neoplasias del tracto respiratorio::neoplasias pulmonares::neoplasias de los bronquios::carcinoma broncogénico::carcinoma de pulmón de células no pequeñas [ENFERMEDADES] ,Protein-Tyrosine Kinases ,phase I ,Good Health and Well Being ,Oncology ,6.1 Pharmaceuticals ,Biochemistry and Cell Biology ,Pulmons - Càncer - Tractament - Abstract
Genomics; Transcriptomics; Lung cancer Genómica; Transcriptómica; Cáncer de pulmón Genòmica; Transcriptòmica; Càncer de pulmó Background The Worldwide Innovative Network (WIN) Consortium has developed the Simplified Interventional Mapping System (SIMS) to better define the cancer molecular milieu based on genomics/transcriptomics from tumor and analogous normal tissue biopsies. SPRING is the first trial to assess a SIMS-based tri-therapy regimen in advanced non-small cell lung cancer (NSCLC). Methods Patients with advanced NSCLC (no EGFR, ALK, or ROS1 alterations; PD-L1 unrestricted; ≤2 prior therapy lines) received avelumab, axitinib, and palbociclib (3 + 3 dose escalation design). Results Fifteen patients were treated (five centers, four countries): six at each of dose levels 1 (DL1) and DL2; three at DL3. The most common ≥Grade 3 adverse events were neutropenia, hypertension, and fatigue. The recommended Phase II dose (RP2D) was DL1: avelumab 10 mg/kg IV q2weeks, axitinib 3 mg po bid, and palbociclib 75 mg po daily (7 days off/21 days on). Four patients (27%) achieved a partial response (PR) (progression-free survival [PFS]: 14, 24, 25 and 144+ weeks), including two after progression on pembrolizumab. Four patients attained stable disease (SD) that lasted ≥24 weeks: 24, 27, 29, and 64 weeks. At DL1 (RP2D), four of six patients (66%) achieved stable disease (SD) ≥6 months/PR (2 each). Responders included patients with no detectable PD-L1 expression and low tumor mutational burden. Conclusions Overall, eight of 15 patients (53%) achieved clinical benefit (SD ≥ 24 weeks/PR) on the avelumab, axitinib, and palbociclib combination. This triplet showed antitumor activity in NSCLC, including in tumors post-pembrolizumab progression, and was active at the RP2D, which was well tolerated. This work was supported by the ARC Foundation for Cancer Research, Villejuif, France and Worldwide Innovative Network (WIN) Association––WIN Consortium, Villejuif, France, sponsor of the study. WIN was responsible for the study design, collection, analysis, and interpretation of data as well as writing of the report. The study drugs were provided by Pfizer, as part of an alliance between Merck KGaA, Darmstadt, Germany and Pfizer. Funded in part by National Cancer Institute grant P30 CA023100 and the Joan and Irwin Jacobs Fund philanthropic fund (RK).
- Published
- 2022
24. Liquid biopsy: current technology and clinical applications
- Author
-
Mina Nikanjam, Shumei Kato, and Razelle Kurzrock
- Subjects
Cancer Research ,Technology ,Oncology ,Liquid Biopsy ,Humans ,Hematology ,Precision Medicine ,Neoplastic Cells, Circulating ,Molecular Biology ,Cell-Free Nucleic Acids ,Immune Checkpoint Inhibitors ,Aged ,Circulating Tumor DNA - Abstract
Liquid biopsies are increasingly used for cancer molecular profiling that enables a precision oncology approach. Circulating extracellular nucleic acids (cell-free DNA; cfDNA), circulating tumor DNA (ctDNA), and circulating tumor cells (CTCs) can be isolated from the blood and other body fluids. This review will focus on current technologies and clinical applications for liquid biopsies. ctDNA/cfDNA has been isolated and analyzed using many techniques, e.g., droplet digital polymerase chain reaction, beads, emulsion, amplification, and magnetics (BEAMing), tagged-amplicon deep sequencing (TAm-Seq), cancer personalized profiling by deep sequencing (CAPP-Seq), whole genome bisulfite sequencing (WGBS-Seq), whole exome sequencing (WES), and whole genome sequencing (WGS). CTCs have been isolated using biomarker-based cell capture, and positive or negative enrichment based on biophysical and other properties. ctDNA/cfDNA and CTCs are being exploited in a variety of clinical applications: differentiating unique immune checkpoint blockade response patterns using serial samples; predicting immune checkpoint blockade response based on baseline liquid biopsy characteristics; predicting response and resistance to targeted therapy and chemotherapy as well as immunotherapy, including CAR-T cells, based on serial sampling; assessing shed DNA from multiple metastatic sites; assessing potentially actionable alterations; analyzing prognosis and tumor burden, including after surgery; interrogating difficult-to biopsy tumors; and detecting cancer at early stages. The latter can be limited by the small amounts of tumor-derived components shed into the circulation; furthermore, cfDNA assessment in all cancers can be confounded by clonal hematopoeisis of indeterminate potential, especially in the elderly. CTCs can be technically more difficult to isolate that cfDNA, but permit functional assays, as well as evaluation of CTC-derived DNA, RNA and proteins, including single-cell analysis. Blood biopsies are less invasive than tissue biopsies and hence amenable to serial collection, which can provide critical molecular information in real time. In conclusion, liquid biopsy is a powerful tool, and remarkable advances in this technology have impacted multiple aspects of precision oncology, from early diagnosis to management of refractory metastatic disease. Future research may focus on fluids beyond blood, such as ascites, effusions, urine, and cerebrospinal fluid, as well as methylation patterns and elements such as exosomes.
- Published
- 2022
25. Tumor Infiltrating Lymphocyte Expression of PD-1 Predicts Response to Anti-PD-1/PD-L1 Immunotherapy
- Author
-
Nicholas J. Bevins, Ryosuke Okamura, Meagan Montesion, Jacob J. Adashek, Aaron M. Goodman, and Razelle Kurzrock
- Subjects
Cancer Research ,Oncology ,Immunology ,Immunology and Allergy - Abstract
Introduction Many studies have focused on the role of programmed death receptor ligand 1 (PD-L1) expression in predicting immunotherapy outcomes. Limited clinical data are available regarding the role of programmed death receptor 1 (PD-1; the PD-L1 receptor) expressing tumor-infiltrating lymphocytes (TILs) in PD-1/PD-L1 antibody responsiveness. However, preclinical studies demonstrate that TILs expressing PD-1 contribute to tumor immune evasion. Methods: This study analyzed the association between TIL-PD-1 status and outcome after immune checkpoint blockade (ICB) therapy. We evaluated 123 patients with various solid tumors treated with monoclonal antibodies targeting the PD-1/PD-L1 signaling axis. Additionally, 8706 solid tumor specimens were assessed for TIL-PD-1 and tumor mutational burden (TMB) status. Results: The presence of PD-1-expressing TILs in tumors was associated with increased median progression-free survival (7.0 vs 1.9 months; p = 0.006) and overall survival (18.1 vs 8.0 months; p = 0.04) after treatment with ICB. TIL-PD-1–positive patients had an objective response rate (ORR) of 41% (95% CI, 24–61; N = 12/29) compared with 17% (95% CI, 4–43; N = 3/17) for TIL-PD-1–negative patients (p = 0.18). Analyzed as continuous variables, TIL-PD-1 and TMB showed a weak correlation in 8706 solid tumor samples (Pearson r = 0.074); when analyzed as categorical variables (cutoffs: TIL-PD-1 ≥ 1% and TMB ≥ 10 mutations/Mb), the two variables are correlated (p < 0.0001). TIL-PD-1–positive status is also associated with enrichment of pathologic variants within several genes, most notably TP53 (adjusted p < 0.05). Conclusion: TIL-PD-1 positivity in tumors (≥ 1%) is associated with significantly longer progression-free and overall survival after ICB. ClinicalTrials.gov ID: NCT02478931
- Published
- 2022
26. Multi-omic analysis in carcinoma of unknown primary (CUP): therapeutic impact of knowing the unknown
- Author
-
Shumei Kato, Sophia Gumas, Jacob J. Adashek, Ryosuke Okamura, Suzanna Lee, Jason K. Sicklick, and Razelle Kurzrock
- Subjects
Cancer Research ,Oncology ,Genetics ,Molecular Medicine ,General Medicine - Abstract
Carcinoma of unknown primary (CUP) is a difficult-to-manage malignancy. Multi-omic profiles and treatment outcome versus degree of precision matching was assessed. Tumors underwent next-generation sequencing (NGS) [tissue and/or blood-derived cell-free DNA (cfDNA)]. Selected patients had transcriptome-based immune profiling and/or programmed cell death 1 ligand 1 (PD-L1) immunohistochemistry analysis. Patients could be reviewed by a Molecular Tumor Board, but physicians chose the therapy. Of 6,497 patients in the precision database, 97 had CUP. The median number of pathogenic tissue genomic alterations was 4 (range 0-25) and, for cfDNA, was 2 (range 0-9). Each patient had a distinct molecular landscape. Food and Drug Administration (FDA)-approved biomarkers included: PD-L1+ ≥1%, 30.9% of CUPs tested; microsatellite instability, 3.6%; tumor mutational burden ≥10 mutations/mb, 23%; neurotrophic receptor tyrosine kinase (NTRK) fusions, 0%. RNA-based immunograms showed theoretically druggable targets: lymphocyte activationgene 3 protein (LAG-3), macrophage colony-stimulating factor 1 receptor (CSF1R), adenosine receptor A2 (ADORA2) and indoleamine2,3-dioxygenase 1 (IDO1). Overall, 56% of patients had ≥1 actionable biomarker (OncoKB database). To quantify the degree of matching (tumors to drugs), a matching score (MS) (roughly equivalent to number of alterations targeted/ total number of deleterious alterations) was calculated post hoc. Comparing evaluable treated patients [MS high,50% (N=15) versus low ≤50% (N=47)], median progression-free survival was 10.4 vs. 2.8 months (95% CI 0.11-0.64; HR 0.27; P=0.002); survival, 15.8 vs. 6.9 months (95% CI 0.17-1.16; HR 0.45; P=0.09); and clinical benefit rate (stable disease ≥6 months/partial/complete response), 71% vs. 24% (P=0.003). Higher MS was the only factor that predicted improvement in outcome variables after multivariate analysis. In conclusion, CUPs are molecularly complex. Treatments with high degrees of matching to molecular alterations (generally achieved by individualized combinations) correlated with improved outcomes.
- Published
- 2022
27. Comprehensive Landscape of Cyclin Pathway Gene Alterations and Co-occurrence with FGF/FGFR Aberrations Across Urinary Tract Tumors
- Author
-
Denis L F Jardim, Sherri Z Millis, Jeffrey S Ross, Scott Lippman, Siraj M Ali, and Razelle Kurzrock
- Subjects
Cancer Research ,Oncology - Abstract
Background Cyclin pathway gene alterations are frequent in urothelial tumors and may co-exist with other important aberrations, leading to therapeutic opportunities. We characterized the landscape of cyclin gene alterations in urothelial and non-urothelial urinary tract (UT) malignancies. Patients and Methods Overall, 6842 urothelial and 897 non-urothelial UT cancers were analyzed (hybrid-capture-based comprehensive genomic profile (Foundation Medicine)). Alteration frequency in cyclin-sensitizing and -resistance genes, and co-occurrence with fibroblast growth factor receptor (FGFR) gene abnormalities were evaluated. Results Cyclin-activating gene alterations were detected in 47.3% of urothelial and 37.9% of non-urothelial UT cancers. Frequency varied by histology and tumor site. CDKN2A and CDKN2B loss were the most frequent alterations in urothelial tumors (present in 38.5% and 30.4% of patients, respectively). Both genes were less frequently altered in adenocarcinomas (15.2% and 8.9%), but commonly altered in squamous cell carcinomas (74.4% and 39%). Tumors of neuroendocrine origin were relatively silent in activating cyclin alterations, but frequently displayed Rb1 alterations (86% and 83.7% of neuroendocrines and small cell carcinomas). Urachal tumors (n = 79) presented a distinct landscape of cyclin alterations relative to other UT cancers, with less frequent alterations overall. FGF/FGFR genes were altered in 34.9% of urothelial (22.1% in FGFR3), and 19.4% of non-urothelial urinary tract tumors (6.8% FGFR3). Cyclin-activating alterations frequently co-occurred with FGF/FGFR alterations but were in general mutually exclusively with cyclin resistance alterations (RB1/CCNE1). Conclusions Cyclin pathway activating alterations are common in urinary tract tumors, but frequency varies with histology and tumors sites. Co-occurrence of cyclin and FGFR pathway alterations may inform therapeutic opportunities.
- Published
- 2022
28. JAK: Not Just Another Kinase
- Author
-
Ruchi P. Agashe, Scott M. Lippman, and Razelle Kurzrock
- Subjects
Cancer Research ,STAT Transcription Factors ,Oncology ,Humans ,COVID-19 ,Janus Kinase Inhibitors ,Tyrosine ,Janus Kinase 1 ,Janus Kinase 2 ,Phosphorylation ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,Janus Kinases - Abstract
The JAK/STAT axis is implicated in cancer, inflammation, and immunity. Numerous cytokines/growth factors affect JAK/STAT signaling. JAKs (JAK1, JAK2, JAK3, and TYK2) noncovalently associate with cytokine receptors, mediate receptor tyrosine phosphorylation, and recruit ≥1 STAT proteins (STAT1, STAT2, STAT3, STAT4, STAT5a, STAT5b, and STAT6). Tyrosine-phosphorylated STATs dimerize and are then transported into the nucleus to function as transcription factors. Signaling is attenuated by specific suppressor of cytokine signaling proteins, creating a negative feedback loop. Both germline mutations and polymorphisms of JAK family members correlate with specific diseases: Systemic lupus erythematosus (TYK2 polymorphisms); severe combined immunodeficiency (JAK3 mutations); pediatric acute lymphoblastic leukemia (TYK2 mutations); and hereditary thrombocytosis (JAK2 mutations). Somatic gain-of-function JAK mutations mainly occur in hematologic malignancies, with the activating JAK2 V617F being a myeloproliferative disorder hallmark; it is also seen in clonal hematopoiesis of indeterminate potential. Several T-cell malignancies, as well as B-cell acute lymphoblastic leukemia, and acute megakaryoblastic leukemia also harbor JAK family somatic alterations. On the other hand, JAK2 copy-number loss is associated with immune checkpoint inhibitor resistance. JAK inhibitors (jakinibs) have been deployed in many conditions with JAK activation; they are approved in myeloproliferative disorders, rheumatoid and psoriatic arthritis, atopic dermatitis, ulcerative colitis, graft-versus-host disease, alopecia areata, ankylosing spondylitis, and in patients hospitalized for COVID-19. Clinical trials are investigating jakinibs in multiple other autoimmune/inflammatory conditions. Furthermore, dermatologic and neurologic improvements have been observed in children with Aicardi–Goutieres syndrome (a genetic interferonopathy) treated with JAK inhibitors.
- Published
- 2022
29. Concomitant MEK and Cyclin Gene Alterations: Implications for Response to Targeted Therapeutics
- Author
-
Ryosuke Okamura, Razelle Kurzrock, Justin Shaya, Jason K. Sicklick, Rebecca E. Jimenez, Shumei Kato, Jacob J. Adashek, and Suzanna Lee
- Subjects
0301 basic medicine ,MAPK/ERK pathway ,Cancer Research ,Palbociclib ,medicine.disease_cause ,Models, Biological ,Cyclin Gene ,03 medical and health sciences ,0302 clinical medicine ,Cyclins ,Neoplasms ,CDKN2B ,Biomarkers, Tumor ,medicine ,Humans ,Genetic Predisposition to Disease ,Molecular Targeted Therapy ,Protein Kinase Inhibitors ,Neoplasm Staging ,Mitogen-Activated Protein Kinase Kinases ,Trametinib ,business.industry ,MEK inhibitor ,Genetic Variation ,Cancer ,Prognosis ,medicine.disease ,Treatment Outcome ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Cancer research ,Disease Susceptibility ,KRAS ,Tomography, X-Ray Computed ,business - Abstract
Purpose: Cyclin and MAPK/MEK-related gene alterations are implicated in cell-cycle progression and cancer growth. Yet, monotherapy to target the cyclin (CDK4/6) or the MEK pathway has often yielded disappointing results. Because coalterations in cyclin and MEK pathway genes frequently cooccur, we hypothesized that resistance to CDK4/6 or MEK inhibitor monotherapy might be mediated via activation of oncogenic codrivers, and that combination therapy might be useful. Experimental Design: Herein, we describe 9 patients with advanced malignancies harboring concomitant CDKN2A and/or CDKN2B alterations (upregulate CDK4/6) along with KRAS or BRAF alterations (activate the MEK pathway) who were treated with palbociclib (CDK4/6 inhibitor) and trametinib (MEK inhibitor) combination-based regimens. Results: Two patients (with pancreatic cancer) achieved a partial remission (PR) and, overall, 5 patients (56%) had clinical benefit (stable disease ≥ 6 months/PR) with progression-free survival of approximately 7, 9, 9, 11, and 17.5+ months. Interestingly, 1 of these patients whose cancer (gastrointestinal stromal tumor) had progressed on MEK targeting regimen, did well for about 1 year after palbociclib was added. Conclusions: These observations suggest that cotargeting cyclin and MEK signaling can be successful when tumors bear genomic coalterations that activate both of these pathways. Further prospective studies using this matching precision strategy to overcome resistance are warranted. See related commentary by Groisberg and Subbiah, p. 2672
- Published
- 2021
- Full Text
- View/download PDF
30. The Impact of COVID-19 on Cancer Clinical Trials Conducted by NCI-Designated Comprehensive Cancer Centers
- Author
-
Diviya Gupta, Razelle Kurzrock, and Shumei Kato
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,2019-20 coronavirus outbreak ,Coronavirus disease 2019 (COVID-19) ,business.industry ,Cancer clinical trial ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,Immunology ,Cancer ,medicine.disease ,Internal medicine ,Immunology and Allergy ,Medicine ,business - Published
- 2020
- Full Text
- View/download PDF
31. Standardized uptake value (SUVmax) in 18F-FDG PET/CT is correlated with the total number of main oncogenic anomalies in cancer patients
- Author
-
Razelle Kurzrock, Amin Haghighat Jahromi, Geraldine Chang, and Carl K. Hoh
- Subjects
0301 basic medicine ,Pharmacology ,Imaging Feature ,Cancer Research ,medicine.medical_specialty ,business.industry ,Radiogenomics ,Standardized uptake value ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Oncology ,030220 oncology & carcinogenesis ,Molecular Medicine ,Medicine ,Fdg pet ct ,Radiology ,business - Abstract
Cancer diagnosis and therapy is quickly moving from the traditional histology-based approaches to genomic stratification, providing a huge opportunity for radiogenomics, associating imaging feature...
- Published
- 2020
- Full Text
- View/download PDF
32. Dual Checkpoint Blockade in a Neuroendocrine Carcinoma With Dual PD-L1/PD-L2 Amplification and High Tumor Mutational Burden
- Author
-
Alexandra Gangi, Jacob J. Adashek, Young Kwang Chae, Jun Gong, Richard Tuli, Sandip Pravin Patel, Gillian Gresham, Michelle Guan, Andrew Eugene Hendifar, Veronica Placencio-Hickok, Razelle Kurzrock, and David P. Frishberg
- Subjects
Cancer Research ,biology ,business.industry ,Case Reports ,DUAL (cognitive architecture) ,Blockade ,Text mining ,Oncology ,PD-L1 ,biology.protein ,Cancer research ,Medicine ,Neuroendocrine carcinoma ,business - Published
- 2020
- Full Text
- View/download PDF
33. Total Number of Alterations in Liquid Biopsies Is an Independent Predictor of Survival in Patients With Advanced Cancers
- Author
-
Yulian Khagi, Aaron M. Goodman, Peter Vu, Razelle Kurzrock, and Paul Riviere
- Subjects
0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,business.industry ,Independent predictor ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Clinical Research ,Circulating tumor DNA ,030220 oncology & carcinogenesis ,Internal medicine ,Original Reports ,Genetics ,medicine ,In patient ,business ,Cancer - Abstract
PURPOSE Studies have demonstrated an association between quantity of circulating tumor DNA (ctDNA) and poorer survival. We investigated the relationship between percent ctDNA (%ctDNA), total number of ctDNA alterations, and overall survival (OS) in liquid biopsies. MATERIALS AND METHODS Overall, 418 patients with blood-based next-generation sequencing (54 to 73 genes) were analyzed. Eligible patients included those who had advanced/metastatic solid tumor malignancies and never received immunotherapy treatment, which may alter the survival curve in patients with high mutational burden. RESULTS Patients with a high (≥ 5%) %ctDNA had significantly shorter OS versus those with intermediate (≥ 0.4% to < 5%) or low (< 0.4%) values (median OS, 7.0 v 14.1 v not reached [NR] months, respectively; P < .0001). Patients with a high (≥ 5) total number of alterations had significantly shorter OS versus those with intermediate (≥ 1.46 to < 5), low (< 1.46), or no alterations (median OS, 4.6 v 11.7 v 21.3 v NR months, respectively; P < .0001). The total number of alterations correlated with %ctDNA (r = 0.85; 95% CI, 0.81 to 0.87; P < .0001). However, only an intermediate to high total number of alterations (≥ 1.46) was an independent predictor of worse OS (hazard ratio, 1.96; 95% CI, 1.30 to 2.96; P = .0014; multivariate analysis). CONCLUSION We demonstrate that the total number of alterations and %ctDNA have prognostic value and correlate with one another, but only the total number of alterations was independently associated with survival outcomes. Our findings suggest that the total number of alterations in plasma may be an indicator of more aggressive tumor biology and therefore poorer survival.
- Published
- 2020
- Full Text
- View/download PDF
34. High Tumor Mutational Burden Correlates with Longer Survival in Immunotherapy-Naïve Patients with Diverse Cancers
- Author
-
Manvita Mareboina, Razelle Kurzrock, Aaron M. Goodman, Theresa J. Whitchurch, Garrett M. Frampton, Andrew B. Sharabi, David Fabrizio, Paul Riviere, Noor Khalid, Rachel Collier, Ryosuke Okamura, Suzanna Lee, Donald A. Barkauskas, and Shumei Kato
- Subjects
Male ,Oncology ,Cancer Research ,medicine.medical_specialty ,medicine.medical_treatment ,Article ,Therapy naive ,Text mining ,Neoplasms ,Internal medicine ,medicine ,Humans ,Survival analysis ,business.industry ,Confounding ,Immunotherapy ,Middle Aged ,medicine.disease ,Survival Analysis ,Primary tumor ,Confidence interval ,Tumor Burden ,Female ,business ,Checkpoint Blockade Immunotherapy - Abstract
Higher tumor mutational burden (TMB) has been correlated with response to checkpoint blockade immunotherapy. However, it is unclear whether TMB independently serves as a prognostic biomarker for outcomes in immunotherapy-naïve patients. Here, we evaluated the relationship between TMB and overall survival in 1,415 immunotherapy-naïve patients with diverse advanced malignancies. TMB was studied both as a tiered variable (low ≤5 mutations/Mb, intermediate >5 and
- Published
- 2020
- Full Text
- View/download PDF
35. Machine learning model to predict oncologic outcomes for drugs in randomized clinical trials
- Author
-
Alexander Schperberg, Razelle Kurzrock, Stéphane Richard, Amélie Boichard, and Igor F. Tsigelny
- Subjects
Cancer Research ,Oncology and Carcinogenesis ,Population ,Datasets as Topic ,Machine learning ,computer.software_genre ,outcome prediction ,law.invention ,Machine Learning ,03 medical and health sciences ,0302 clinical medicine ,Randomized controlled trial ,law ,Neoplasms ,Antineoplastic Combined Chemotherapy Protocols ,Biomarkers, Tumor ,medicine ,Humans ,Oncology & Carcinogenesis ,drug-related biomarkers ,Lung cancer ,education ,Randomized Controlled Trials as Topic ,clinical trials ,education.field_of_study ,Models, Genetic ,business.industry ,Clinical study design ,Cancer ,Prognosis ,medicine.disease ,Progression-Free Survival ,Clinical trial ,machine learning ,Oncology ,Drug Resistance, Neoplasm ,Research Design ,030220 oncology & carcinogenesis ,Mutation ,Adenocarcinoma ,Biomarker (medicine) ,Artificial intelligence ,molecular profiles ,business ,computer ,Forecasting - Abstract
Predicting oncologic outcome is challenging due to the diversity of cancer histologies and the complex network of underlying biological factors. In this study, we determine whether machine learning (ML) can extract meaningful associations between oncologic outcome and clinical trial, drug-related biomarker and molecular profile information. We analyzed therapeutic clinical trials corresponding to 1102 oncologic outcomes from 104 758 cancer patients with advanced colorectal adenocarcinoma, pancreatic adenocarcinoma, melanoma and nonsmall-cell lung cancer. For each intervention arm, a dataset with the following attributes was curated: line of treatment, the number of cytotoxic chemotherapies, small-molecule inhibitors, or monoclonal antibody agents, drug class, molecular alteration status of the clinical arm's population, cancer type, probability of drug sensitivity (PDS) (integrating the status of genomic, transcriptomic and proteomic biomarkers in the population of interest) and outcome. A total of 467 progression-free survival (PFS) and 369 overall survival (OS) data points were used as training sets to build our ML (random forest) model. Cross-validation sets were used for PFS and OS, obtaining correlation coefficients (r) of 0.82 and 0.70, respectively (outcome vs model's parameters). A total of 156 PFS and 110 OS data points were used as test sets. The Spearman correlation (rs ) between predicted and actual outcomes was statistically significant (PFS: rs = 0.879, OS: rs = 0.878, P
- Published
- 2020
- Full Text
- View/download PDF
36. Role of ultraviolet mutational signature versus tumor mutation burden in predicting response to immunotherapy
- Author
-
Paul Riviere, Amélie Boichard, Pablo Tamayo, Razelle Kurzrock, Timothy V. Pham, Huwate Yeerna, and Aaron M. Goodman
- Subjects
0301 basic medicine ,Male ,Cancer Research ,Ultraviolet Rays ,medicine.medical_treatment ,Kaplan-Meier Estimate ,Major histocompatibility complex ,lcsh:RC254-282 ,UV mutational signature ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Antigens, Neoplasm ,Genetics ,medicine ,Humans ,In patient ,Exome ,Amino Acids ,Research Articles ,biology ,business.industry ,Genome, Human ,Immunogenicity ,Melanoma ,General Medicine ,Immunotherapy ,Middle Aged ,medicine.disease ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Progression-Free Survival ,Blockade ,Tumor Burden ,Immune recognition ,030104 developmental biology ,Oncology ,Mutagenesis ,030220 oncology & carcinogenesis ,Multivariate Analysis ,Mutation ,biology.protein ,Cancer research ,Molecular Medicine ,checkpoint blockade ,Female ,immunotherapy ,business ,Hydrophobic and Hydrophilic Interactions ,Research Article - Abstract
Hydrophobic neoantigens are more immunogenic because they are better presented by the major histocompatibility complex and better recognized by T cells. Tumor cells can evade the immune response by expressing checkpoints such as programmed death ligand 1. Checkpoint blockade reactivates immune recognition and can be effective in diseases such as melanoma, which harbors a high tumor mutational burden (TMB). Cancers presenting low or intermediate TMB can also respond to checkpoint blockade, albeit less frequently, suggesting the need for biological markers predicting response. We calculated the hydrophobicity of neopeptides produced by probabilistic in silico simulation of the genomic UV exposure mutational signature. We also computed the hydrophobicity of potential neopeptides and extent of UV exposure based on the UV mutational signature enrichment (UVMSE) score in The Cancer Genome Atlas (TCGA; N = 3543 tumors), and in our cohort of 151 immunotherapy‐treated patients. In silico simulation showed that UV exposure significantly increased hydrophobicity of neopeptides, especially over multiple mutagenic cycles. There was also a strong correlation (R 2 = 0.953) between weighted UVMSE and hydrophobicity of neopeptides in TCGA melanoma patients. Importantly, UVMSE was able to predict better response (P = 0.0026), progression‐free survival (P = 0.036), and overall survival (P = 0.052) after immunotherapy in patients with low/intermediate TMB, but not in patients with high TMB. We show that higher UVMSE scores could be a useful predictor of better immunotherapy outcome, especially in patients with low/intermediate TMB, likely due to increased hydrophobicity (and hence immunogenicity) of neopeptides., The mutational pattern induced by ultraviolet radiation changes the exome to encode for more hydrophobic antigens. When these more lipophilic antigens are presented on the cell surface to T cells by major histocompatibility complex I molecules, they could induce a stronger immune response. This was demonstrated in a cohort of 151 patients from the Moores Cancer Center, particularly in the low TMB subset.
- Published
- 2020
37. Angiosarcoma heterogeneity and potential therapeutic vulnerability to immune checkpoint blockade: insights from genomic sequencing
- Author
-
Michael J. Wagner, Razelle Kurzrock, and Amélie Boichard
- Subjects
lcsh:QH426-470 ,medicine.medical_treatment ,Hemangiosarcoma ,lcsh:Medicine ,Personalized therapy ,Biology ,Genetic Heterogeneity ,Genetics ,medicine ,Biomarkers, Tumor ,Angiosarcoma ,Missense mutation ,Humans ,Genetic Predisposition to Disease ,Molecular Targeted Therapy ,Precision Medicine ,Molecular Biology ,Gene ,Immune Checkpoint Inhibitors ,neoplasms ,Genetics (clinical) ,Melanoma ,Research ,lcsh:R ,Immunotherapy ,medicine.disease ,Human genetics ,Immune checkpoint ,digestive system diseases ,Blockade ,lcsh:Genetics ,Treatment Outcome ,Mutagenesis ,Mutation ,Cancer research ,Molecular Medicine ,Disease Susceptibility - Abstract
Background Angiosarcoma is an aggressive tumor. Recent case series describe exceptional responses to checkpoint blockade in this disease. Methods Herein, we explored the genomic correlates of 48 angiosarcomas from the Angiosarcoma Project (12,499 variants analyzed in 6603 genes; whole-exome sequencing) versus 10,106 pan-cancer tumors in The Cancer Genome Atlas including 235 sarcomas but no angiosarcoma. Results At the molecular level, angiosarcomas were heterogeneous. Those located in the face and scalp presented high tumor mutation burden, missense amino acid variations biased towards more hydrophobic (and therefore more immunogenic) peptides, and ultra-violet mutational signature. Conclusions Angiosarcoma molecular features are similar to those observed in melanoma and other skin tumors and may explain comparable immunotherapy sensitivity of these tumor types.
- Published
- 2020
- Full Text
- View/download PDF
38. Precision oncology: the intention-to-treat analysis fallacy
- Author
-
Ryosuke Okamura, Jason K. Sicklick, Shumei Kato, and Razelle Kurzrock
- Subjects
0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,Clinical Trials and Supportive Activities ,Oncology and Carcinogenesis ,Medical Oncology ,Article ,law.invention ,03 medical and health sciences ,0302 clinical medicine ,Bias ,Randomized controlled trial ,Clinical Research ,law ,Neoplasms ,Internal medicine ,Humans ,Medicine ,False Positive Reactions ,Molecular Targeted Therapy ,Oncology & Carcinogenesis ,Precision Medicine ,Protein Kinase Inhibitors ,Cancer ,Randomized Controlled Trials as Topic ,Response rate (survey) ,Intention-to-treat analysis ,business.industry ,Confounding ,Evaluation of treatments and therapeutic interventions ,Precision oncology ,Precision medicine ,Intention to Treat Analysis ,Clinical trial ,Good Health and Well Being ,Treatment Outcome ,030104 developmental biology ,Intention-to-treat ,5.1 Pharmaceuticals ,Research Design ,6.1 Pharmaceuticals ,030220 oncology & carcinogenesis ,Public Health and Health Services ,Biomarker (medicine) ,Development of treatments and therapeutic interventions ,business - Abstract
It has recently been suggested that precision oncology studies should be reanalysed using the intention-to-treat (ITT) methodology developed for randomized controlled clinical trials. This reanalysis dramatically decreases response rates in precision medicine studies. We contend that the ITT analysis of precision oncology trials is invalid. The ITT methodology was developed three decades ago to mitigate the problems of randomized trials, which try to ensure that both arms have an unselected patient population free from confounders. In contrast, precision oncology trials specifically select patients for confounders (that is biomarkers) that predict response. To demonstrate the issues inherent in an ITT reanalysis for precision cancer medicine studies, we take as an example the drug larotrectinib (TRK inhibitor) approved because of remarkable responses in malignancies harbouring NTRK fusions. Based on large-scale studies, NTRK fusions are found in ~0.31% of tumours. In a non-randomized pivotal study of larotrectinib, 75% of the 55 treated patients responded. Based upon the prevalence of NTRK fusions, ~18,000 patients would need to be screened to enrol the 55 treated patients. Utilizing the ITT methodology, the revised response rate to larotrectinib would be 0.23%. This is, of course, a dramatic underestimation of the efficacy of this now Food and Drug Administration (FDA)-approved drug. Similar issues can be shown for virtually any biomarker-based precision clinical trial. Therefore, retrofitting the ITT analysis developed for unselected patient populations in randomized trials yields misleading conclusions in precision medicine studies.
- Published
- 2020
- Full Text
- View/download PDF
39. Relationship between protein biomarkers of chemotherapy response and microsatellite status, tumor mutational burden and PD‐L1 expression in cancer patients
- Author
-
Zoran Gatalica, Mina Nikanjam, Donald A. Barkauskas, Jeff Swensen, Razelle Kurzrock, and David Arguello
- Subjects
PD-L1 ,Cancer Research ,medicine.medical_treatment ,Dacarbazine ,Oncology and Carcinogenesis ,Programmed Cell Death 1 Receptor ,Antineoplastic Agents ,Thymidylate synthase ,B7-H1 Antigen ,03 medical and health sciences ,0302 clinical medicine ,Neoplasms ,cytotoxic chemotherapy ,Biomarkers, Tumor ,medicine ,Humans ,Oncology & Carcinogenesis ,Cancer Therapy and Prevention ,MSI ,Etoposide ,Chemotherapy ,Temozolomide ,biology ,TMB ,business.industry ,High-Throughput Nucleotide Sequencing ,Gemcitabine ,Oncology ,PD‐L1 ,030220 oncology & carcinogenesis ,Cancer research ,biology.protein ,Female ,Microsatellite Instability ,immunotherapy ,ERCC1 ,business ,Microsatellite Repeats ,medicine.drug ,Epirubicin - Abstract
Chemotherapy and checkpoint inhibitor immunotherapies are increasingly used in combinations. We determined associations between the presence of anti‐PD‐1/PD‐L1 therapeutic biomarkers and protein markers of potential chemotherapy response. Data were extracted from a clinical‐grade testing database (Caris Life Sciences; February 2015 through November 2017): immunotherapy response markers (microsatellite instability‐high [MSI‐H], tumor mutational burden‐high [TMB‐H], and PD‐L1 protein expression) and protein chemotherapy response markers (excision repair complementation group 1 [ERCC1], topoisomerase 1 [TOPO1], topoisomerase 2 [TOP2A], thymidylate synthase [TS], tubulin beta 3 [TUBB3], ribonucleotide reductase regulatory subunit M1 [RRM1] and O‐6‐methyl guanine DNA methyltransferase [MGMT]). Relationships were determined by the Mantel‐Haenszel chi‐squared test or Fischer's exact tests. Overall, 28,034 patients representing a total of 40 tumor types were assessed. MSI‐H was found in 3.3% of patients (73% were also TMB‐H), TMB‐H, 8.4% (28.3% were also MSI‐H) and PD‐L1 expression in 11.0% of patients (5.1% were also MSI‐H; 16.4% were also TMB‐H). Based on concurrent biomarker expression, combinations of immunotherapy with platinum (ERCC1 negativity) or with doxorubicin, epirubicin or etoposide (TOP2A positivity) have a higher probability of response, whereas combinations with irinotecan or topotecan (TOPO1 positivity), with gemcitabine (RRM1 negativity), and fluorouracil, pemetrexed or capecitabine (TS negativity) may be of less benefit. The potential for immunotherapy and taxane (TUBB3 negativity) combinations is present for MSI‐H but not TMB‐H or PD‐L1‐expressing tumors; for temozolomide and dacarbazine (MGMT negative), PD‐L1 is frequently coexpressed, but MSI‐H and TMB‐H are not associated. Protein markers of potential chemotherapy response along with next‐generation sequencing for immunotherapy response markers can help support rational combinations as part of an individualized, precision oncology approach., What's new? With the emerging success of immunotherapy of cancers, combinations with conventional chemotherapies are increasingly being tested in clinical trials. Here the authors examined concurrent biomarker expression of checkpoint (PD‐1/PD‐L1) blockade immunotherapy and various cytotoxic chemotherapies to determine which chemotherapeutic agents will best synergize with immunotherapy. They predict that combining platinum or doxorubicin, epirubicin, or etoposide treatments with PD‐1/PD‐L1 inhibitors would have a higher probability of response than other treatments, supporting a rational combination strategy in a possibly individualized treatment approach.
- Published
- 2020
- Full Text
- View/download PDF
40. A Phase II Basket Trial of Dual Anti–CTLA-4 and Anti–PD-1 Blockade in Rare Tumors (DART SWOG 1609) in Patients with Nonpancreatic Neuroendocrine Tumors
- Author
-
Annette Fontaine, Anup Kasi, Jourdain Hayward, Tareq Al Baghdadi, Manisha Shah, Melissa Plets, Megan Othus, Razelle Kurzrock, Edward Mayerson, Zoran Gatalica, Francis J. Giles, Christopher W. Ryan, Donna E. Hansel, Christine M. McLeod, Sandip Pravin Patel, Elad Sharon, Helen X. Chen, Marc Matrana, W. Michael Korn, Preet Paul Singh, Young Kwang Chae, and Charles D. Blanke
- Subjects
Male ,0301 basic medicine ,Cancer Research ,Programmed Cell Death 1 Receptor ,Phases of clinical research ,Neuroendocrine tumors ,Gastroenterology ,0302 clinical medicine ,Antineoplastic Combined Chemotherapy Protocols ,80 and over ,Clinical endpoint ,CTLA-4 Antigen ,Prospective Studies ,Prospective cohort study ,Cancer ,Aged, 80 and over ,Middle Aged ,Prognosis ,Survival Rate ,Neuroendocrine Tumors ,Nivolumab ,Oncology ,6.1 Pharmaceuticals ,030220 oncology & carcinogenesis ,Female ,medicine.drug ,Adult ,medicine.medical_specialty ,Clinical Trials and Supportive Activities ,Oncology and Carcinogenesis ,Ipilimumab ,Article ,Pancreatic Cancer ,03 medical and health sciences ,Rare Diseases ,Clinical Research ,Internal medicine ,medicine ,Humans ,Oncology & Carcinogenesis ,Intermediate Grade ,Survival rate ,Aged ,business.industry ,Neurosciences ,Evaluation of treatments and therapeutic interventions ,medicine.disease ,030104 developmental biology ,Digestive Diseases ,business ,Follow-Up Studies - Abstract
Purpose: Immune checkpoint blockade has improved outcomes across tumor types; little is known about the efficacy of these agents in rare tumors. We report the results of the (nonpancreatic) neuroendocrine neoplasm cohort of SWOG S1609 dual anti–CTLA-4 and anti–PD-1 blockade in rare tumors (DART). Patients and Methods: We performed a prospective, open-label, multicenter phase II clinical trial of ipilimumab plus nivolumab across multiple rare tumor cohorts, with the (nonpancreatic) neuroendocrine cohort reported here. Response assessment by grade was not prespecified. The primary endpoint was overall response rate [ORR; RECIST v1.1; complete response (CR) and partial response (PR)]; secondary endpoints included progression-free survival (PFS), overall survival (OS), stable disease >6 months, and toxicity. Results: Thirty-two eligible patients received therapy; 18 (56%) had high-grade disease. Most common primary sites were gastrointestinal (47%; N = 15) and lung (19%; N = 6). The overall ORR was 25% [95% confidence interval (CI) 13–64%; CR, 3%, N = 1; PR, 22%, N = 7]. Patients with high-grade neuroendocrine carcinoma had an ORR of 44% (8/18 patients) versus 0% in low/intermediate grade tumors (0/14 patients; P = 0.004). The 6-month PFS was 31% (95% CI, 19%–52%); median OS was 11 months (95% CI, 6–∞). The most common toxicities were hypothyroidism (31%), fatigue (28%), and nausea (28%), with alanine aminotransferase elevation (9%) as the most common grade 3/4 immune-related adverse event, and no grade 5 events. Conclusions: Ipilimumab plus nivolumab demonstrated a 44% ORR in patients with nonpancreatic high-grade neuroendocrine carcinoma, with 0% ORR in low/intermediate grade disease.
- Published
- 2020
- Full Text
- View/download PDF
41. Concordance between TP53 alterations in blood and tissue: impact of time interval, biopsy site, cancer type and circulating tumor DNA burden
- Author
-
Ryosuke Okamura, Razelle Kurzrock, and Cheyennedra C. Bieg-Bourne
- Subjects
Male ,0301 basic medicine ,Cancer Research ,Time Factors ,Colorectal cancer ,Biopsy ,Gastroenterology ,Circulating Tumor DNA ,0302 clinical medicine ,Biopsy Site ,Neoplasms ,TP53 ,Neoplasm Metastasis ,Research Articles ,Aged, 80 and over ,General Medicine ,Middle Aged ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Oncology ,Circulating tumor DNA ,030220 oncology & carcinogenesis ,Molecular Medicine ,Female ,Research Article ,Tissue biopsy ,Adult ,concordance ,medicine.medical_specialty ,Concordance ,Context (language use) ,lcsh:RC254-282 ,Young Adult ,03 medical and health sciences ,Internal medicine ,genomics ,Genetics ,medicine ,Humans ,cancer ,Aged ,Genome, Human ,business.industry ,Cancer type ,Cancer ,ctDNA ,medicine.disease ,030104 developmental biology ,Genetic Loci ,Tumor Suppressor Protein p53 ,business - Abstract
We examined the impact of spatial, temporal, histologic, and quantitative factors on concordance between TP53 alterations in tissue DNA vs in circulating tumor DNA (ctDNA). Four hundred and thirty‐three patients underwent next‐generation sequencing (NGS) in which both tissue and blood samples were evaluated. TP53 was detected in 258 of 433 patients (59.6%); 215 had tissue TP53 alterations (49.7%); 159, ctDNA (36.7%); and 116, both tissue and ctDNA (27.8%). Overall concordance rate between ctDNA and tissue biopsies for TP53 alterations was 67.2%; positive concordance was 45.0%. Overall concordance for TP53 did not vary among patients with ≤ 2 months vs > 6 months between test samples; however, positive concordance trended higher when time intervals between test samples were shorter, suggesting that the lack of difference in overall concordance may be due to the large number of negative/negative tests. There was a trend toward higher overall concordance based on biopsy site (metastatic vs primary) (P = 0.07) and significantly higher positive concordance if the tissue biopsy site was a metastatic lesion (P = 0.03). Positive concordance significantly decreased in noncolorectal cancer patients vs colorectal cancer patients (P = 0.02). Finally, higher %ctDNA was associated with higher concordance rates between blood and tissue (P, This study presents evidence that the evaluation of the full spectrum of TP53 alterations in patients with diverse malignancies requires sequencing of both blood‐derived circulating tumor DNA and tissue DNA. Multiple factors can influence positive concordance rates between the tests, including site of tissue biopsy, %ctDNA, time between tissue biopsy and blood sample, and histologic context.
- Published
- 2020
- Full Text
- View/download PDF
42. Dual EGFR blockade with cetuximab and erlotinib combined with anti-VEGF antibody bevacizumab in advanced solid tumors: a phase 1 dose escalation triplet combination trial
- Author
-
Yunfang Jiang, Vivek Subbiah, David S. Hong, Apostolia Maria Tsimberidou, Filip Janku, Gerald Steven Falchook, Razelle Kurzrock, Kyaw Zin Thein, Aung Naing, Ecaterina Ileana Dumbrava, Funda Meric-Bernstam, Siqing Fu, and Sarina Anne Piha-Paul
- Subjects
0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,Bevacizumab ,Phase 1 dose escalation ,lcsh:RC254-282 ,Metastasis ,03 medical and health sciences ,0302 clinical medicine ,Epidermal growth factor ,Internal medicine ,medicine ,Hematology ,Cetuximab ,business.industry ,lcsh:RC633-647.5 ,Research ,Cancer ,Cetuximab, erlotinib and bevacizumab ,lcsh:Diseases of the blood and blood-forming organs ,medicine.disease ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Dual EGFR blockade ,030104 developmental biology ,Response Evaluation Criteria in Solid Tumors ,030220 oncology & carcinogenesis ,Erlotinib ,Advanced solid tumors ,business ,medicine.drug - Abstract
Background Angiogenesis and activation of the epidermal growth factor (EGFR) pathway play an essential role in tumor proliferation and metastasis. Targeting angiogenesis or EGFR alone does not yield adequate tumor control in most solid tumors. Overcoming intrinsic and/or acquired resistance may need a doublet or triplet therapy strategy. Herein, we report the safety and feasibility of dual EGFR blockade with EGFR monoclonal antibody and EGFR tyrosine kinase inhibitor combined with anti-VEGF antibody in advanced solid tumors. Methods We conducted a phase I study combining erlotinib, cetuximab, and bevacizumab. Patients with advanced or metastatic solid tumors (excluding colorectal and non-small cell lung cancers) were analyzed for safety, toxicity profile, and response. Anti-tumor activity was evaluated per response evaluation criteria in solid tumors (RECIST 1.0). Results Thirty-six patients received treatment on a range of dose-levels. The most frequent tumor types enrolled were cervical (n = 10), head and neck squamous cell (n = 10), and follicular thyroid (n = 4) cancers. The most common treatment-related grade ≥ 2 adverse events were rash (56%), hypomagnesemia (17%), pruritus (11%), diarrhea (8%), and tumor-related bleeding (8%). Seventeen of 19 patients (89%) treated at the maximum tolerated dose did not present treatment-related dose-limiting toxicity. Fifteen (63%) of the 24 evaluable patients achieved a disease control (stable disease ≥ 4 months (n = 14) and partial response (n = 1). The median number of prior lines of therapies was 3 (range 1–10). Conclusions The triplet combination of erlotinib, cetuximab, and bevacizumab was well tolerated, conferring clinical benefit in heavily pretreated patients. Future studies are warranted with second or third-generation EGFR tyrosine kinase triplet combinations in the EGFR pathway aberrant patients. Trial Registration: ClinicalTrials.gov Identifier: NCT00543504. Sponsor(s): National Cancer Institute (NCI), MD Anderson Cancer Center
- Published
- 2020
- Full Text
- View/download PDF
43. Development of 2 Bromodomain and Extraterminal Inhibitors With Distinct Pharmacokinetic and Pharmacodynamic Profiles for the Treatment of Advanced Malignancies
- Author
-
Julie Switzky, Gerald Steven Falchook, Monica M. Mita, Russell Z. Szmulewitz, Swamy Yeleswaram, Wael A. Harb, Moshe Talpaz, Shilpa Gupta, Razelle Kurzrock, Justin M. Watts, Julio Antonio Peguero, Gongfu Zhou, Phillip Liu, Amitkumar Mehta, Marcus Smith Noel, Seth Rosen, Ryan D. Cassaday, Patricia LoRusso, Catherine C. Coombs, Fred Zheng, Sarina Anne Piha-Paul, and David Smith
- Subjects
Male ,0301 basic medicine ,Oncology ,Cancer Research ,0302 clinical medicine ,Neoplasms ,80 and over ,Tissue Distribution ,Organic Chemicals ,Aged, 80 and over ,Nausea ,Middle Aged ,Boronic Acids ,Treatment Outcome ,Tolerability ,6.1 Pharmaceuticals ,030220 oncology & carcinogenesis ,Female ,Patient Safety ,Drug ,medicine.symptom ,Adult ,medicine.medical_specialty ,Maximum Tolerated Dose ,Adolescent ,Vomiting ,Clinical Trials and Supportive Activities ,Oncology and Carcinogenesis ,Antineoplastic Agents ,Article ,Dose-Response Relationship ,Young Adult ,03 medical and health sciences ,Therapeutic index ,Pharmacokinetics ,Clinical Research ,Internal medicine ,medicine ,Humans ,Oncology & Carcinogenesis ,Dosing ,Adverse effect ,Aged ,Dose-Response Relationship, Drug ,business.industry ,Proteins ,Evaluation of treatments and therapeutic interventions ,Clinical trial ,Pyrimidines ,030104 developmental biology ,Pharmacodynamics ,business - Abstract
Purpose: Bromodomain and extraterminal (BET) proteins are key epigenetic transcriptional regulators, inhibition of which may suppress oncogene expression. We report results from 2 independent first-in-human phase 1/2 dose–escalation and expansion, safety and tolerability studies of BET inhibitors INCB054329 (study INCB 54329-101; NCT02431260) and INCB057643 (study INCB 57643-101; NCT02711137). Patients and Methods: Patients (≥18 years) with advanced malignancies, ≥1 prior therapy, and adequate organ functions received oral INCB054329 (monotherapy) or INCB057643 (monotherapy or in combination with standard-of-care) in 21-day cycles (or 28-day cycles depending on standard-of-care combination). Primary endpoints were safety and tolerability. Results: Sixty-nine and 134 patients received INCB054329 and INCB057643, respectively. Study INCB 54329-101 has been completed; INCB 57643-101 is currently active, but not recruiting (no patients were receiving treatment as of January 8, 2019). Terminal elimination half-life was shorter for INCB054329 versus INCB057643 (mean [SD], 2.24 [2.03] vs. 11.1 [8.27] hours). INCB054329 demonstrated higher interpatient variability in oral clearance versus INCB057643 (CV%, 142% vs. 45.5%). Most common (>20%) any-grade treatment-related adverse events were similar for both drugs (INCB054329; INCB057643): nausea (35%; 30%), thrombocytopenia (33%; 32%), fatigue (29%; 30%), decreased appetite (26%; 22%). Two confirmed complete responses and 4 confirmed partial responses with INCB057643 were reported as best responses. Conclusions: INCB057643 exhibited a more favorable PK profile versus INCB054329; exposure-dependent thrombocytopenia was observed with both drugs which limited the target inhibition that could be safely maintained. Further efforts are required to identify patient populations that can benefit most, and an optimal dosing scheme to maximize therapeutic index.
- Published
- 2020
- Full Text
- View/download PDF
44. Phase I dose-escalation trial of the oral AKT inhibitor uprosertib in combination with the oral MEK1/MEK2 inhibitor trametinib in patients with solid tumors
- Author
-
Cathrine Leonowens, Theresa L. Werner, Rebecca S. Heist, Anthony D'Amelio, Li Yan, Rene Gonzalez, Jennifer Gauvin, Carlos Becerra, Joseph F. Kleha, Catherine E. Ellis, Chenxi Wang, Nageatte Ibrahim, Shanker Kalyana-Sundaram, Anthony W. Tolcher, Razelle Kurzrock, V. Valero, Julie Means-Powell, and Antoinette R. Tan
- Subjects
0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,Toxicology ,03 medical and health sciences ,0302 clinical medicine ,Breast cancer ,Internal medicine ,medicine ,Pharmacology (medical) ,Dosing ,Adverse effect ,Pharmacology ,Trametinib ,business.industry ,MEK inhibitor ,medicine.disease ,Rash ,030104 developmental biology ,Tolerability ,030220 oncology & carcinogenesis ,Toxicity ,medicine.symptom ,business - Abstract
This study aimed to determine the safety, tolerability, and recommended phase II doses of trametinib plus uprosertib (GSK2141795) in patients with solid tumors likely to be sensitive to MEK and/or AKT inhibition. This was a phase I, open-label, dose-escalation, and dose-expansion study in patients with triple-negative breast cancer or BRAF-wild type advanced melanoma. The primary outcome of the expansion study was investigator-assessed response. Among 126 enrolled patients, 63 received continuous oral daily dosing of trametinib and uprosertib, 29 received various alternative dosing schedules, and 34 were enrolled into expansion cohorts. Doses tested in the expansion cohort were trametinib 1.5 mg once daily (QD) + uprosertib 50 mg QD. Adverse events (AEs) were consistent with those reported in monotherapy studies but occurred at lower doses and with greater severity. Diarrhea was the most common dose-limiting toxicity; diarrhea and rash were particularly difficult to tolerate. Overall, 59% and 6% of patients reported AEs with a maximum severity of grade 3 and 4, respectively. Poor tolerability prevented adequate delivery of uprosertib with trametinib at a concentration predicted to have clinical activity. The study was terminated early based on futility in the continuous-dosing expansion cohorts and a lack of pharmacological or therapeutic advantage with intermittent dosing. The objective response rate was
- Published
- 2020
- Full Text
- View/download PDF
45. Case series of outcomes in advanced cancer patients with single pathway alterations receiving N-of-One therapies
- Author
-
Diviya Gupta, Razelle Kurzrock, Suzanna Lee, Ryosuke Okamura, Hyo Jeong Lim, Ki Hwan Kim, Jason K. Sicklick, and Shumei Kato
- Subjects
Cancer Research ,Oncology - Abstract
Though advanced cancers generally display complex molecular portfolios, there is a subset of patients whose malignancies possess only one genomic alteration or alterations in one oncogenic pathway. We assess how N-of-One therapeutic strategies impact outcomes in these patients. From 12/2012 to 9/2018, 429 therapy-evaluable patients with diverse treatment-refractory cancers were presented at Molecular Tumor Boards at Moores Cancer Center at UC San Diego. The clinical benefit rate, defined by RECIST1.1, was assessed for patients with solid tumors who underwent next-generation sequencing (NGS) profiling revealing one genomic or pathway alteration, subsequently managed with N-of-One therapies. Nine of 429 patients (2.1%) met evaluation criteria. Using matched therapy indicated by NGS, the clinical benefit rate (stable disease ≥ 6 months/partial/complete response) was 66.7%. Median progression-free survival was 11.3 months (95% CI: 3.4–not evaluable). Thus, a small subset of diverse cancers has single pathway alterations on NGS testing. These patients may benefit from customized therapeutic matching.
- Published
- 2022
- Full Text
- View/download PDF
46. Association of CD274 (PD-L1) Copy Number Changes with Immune Checkpoint Inhibitor Clinical Benefit in Non-Squamous Non-Small Cell Lung Cancer
- Author
-
Karthikeyan Murugesan, Dexter X Jin, Leah A Comment, David Fabrizio, Priti S Hegde, Julia A Elvin, Brian Alexander, Mia A Levy, Garrett M Frampton, Meagan Montesion, Sameek Roychowdhury, Razelle Kurzrock, Jeffrey S Ross, Lee A Albacker, and Richard S P Huang
- Subjects
Cancer Research ,Lung Neoplasms ,Oncology ,DNA Copy Number Variations ,Carcinoma, Non-Small-Cell Lung ,Humans ,Prospective Studies ,Immune Checkpoint Inhibitors ,B7-H1 Antigen - Abstract
Background We sought to characterize response to immune checkpoint inhibitor (ICI) in non-squamous non-small cell lung cancer (NSCLC) across various CD274 copy number gain and loss thresholds and identify an optimal cutoff. Materials and Methods A de-identified nationwide (US) real-world clinico-genomic database was leveraged to study 621 non-squamous NSCLC patients treated with ICI. All patients received second-line ICI monotherapy and underwent comprehensive genomic profiling as part of routine clinical care. Overall survival (OS) from start of ICI, for CD274 copy number gain and loss cohorts across varying copy number thresholds, were assessed. Results Among the 621 patients, patients with a CD274 CN greater than or equal to specimen ploidy +2 (N = 29) had a significantly higher median (m) OS when compared with the rest of the cohort (N = 592; 16.1 [8.9-37.3] vs 8.6 [7.1-10.9] months, hazard ratio (HR) = 0.6 [0.4-1.0], P-value = .05). Patients with a CD274 copy number less than specimen ploidy (N = 299) trended toward a lower mOS when compared to the rest of the cohort (N = 322; 7.5 [5.9-11.3] vs 9.6 [7.9-12.8] months, HR = 0.9 [0.7-1.1], P-value = .3). Conclusion This work shows that CD274 copy number gains at varying thresholds predict different response to ICI blockade in non-squamous NSCLC. Considering these data, prospective clinical trials should further validate these findings, specifically in the context of PD-L1 IHC test results.
- Published
- 2022
47. Abstract CT161: A phase II basket trial of dual anti-CTLA-4 and anti-PD-1 blockade in rare tumors (DART) SWOG S1609: the non-epithelial ovarian tumor cohort
- Author
-
Young Kwang Chae, Megan Othus, Sandip Pravin Patel, Kelly J. Wilkinson, Emily M. Whitman-Purves, Jayanthi Lea, John M. Schallenkamp, Nabil Adra, Leonard J. Appleman, Mitchell Alden, Jessica Thomes Pepin, John A. Ellerton, Andrew Poklepovic, Adam Walter, Murtuza M. Rampurwala, William R. Robinson, Liam Il-Young Chung, Christine M. McLeod, Helen X. Chen, Elad Sharon, Howard Streicher, Christopher W. Ryan, Charles D. Blanke, and Razelle Kurzrock
- Subjects
Cancer Research ,Oncology - Abstract
Background: Dual checkpoint inhibition with Anti-PD-1 and anti-CTLA4 checkpoint inhibitors have proven to be effective in several malignancies but their potential role in numerous rare solid cancers is yet to be established. This study presents the first results of ipilimumab and nivolumab in the non-epithelial ovarian tumor cohort (#13) of the SWOG S1609 Dual Anti-CTLA-4 & Anti-PD-1 blockade in Rare Tumors (DART) trial. Methods: DART is a prospective, open-label, multicenter/multi-cohort phase 2 clinical trial of ipilimumab (1mg/kg intravenously every 6 weeks) plus nivolumab (240mg intravenously every 2 weeks). This cohort included several histologies grouped for statistical analysis. The primary endpoint was objective response rate (ORR) (RECIST v1.1) (confirmed complete (CR) and partial responses (PR)); progression-free survival (PFS), overall survival (OS), stable disease (SD) >6 months, and toxicity are secondary endpoints. Results: Seventeen evaluable patients (median age, 64 years) were analyzed. The subtypes of non-epithelial ovarian cancer were: granulosa cell (47%, n=8), carcinosarcoma or malignant mixed Mullerian tumor (MMMT; 35%, n=6), one each for Wolffian duct, yolk sac, and Sertoli-Leydig cell. There were 2 responses in the 8 patients with granulosa cell (ORR of 25% in the granulosa cell tumors): 1CR (79% regression [due to lymph node < 1.0cm], 59 month PFS, juvenile type) and 1 PR (79% regression, 51+ month PFS, adult type). 6/8 patients remain alive (PFS 52-1774 days). In contrast, carcinosarcomas showed no responses. One patient with Sertoli-Leydig cell tumor had a 22% response and 341 day PFS. Overall ORR was 12% (2/17), clinical benefit rate (CBR; no progression > 6months) of 29.4%. The median PFS was 3.5 months, median OS was 42.5 months. The most common adverse events were fatigue (52.9%, n=9) and hypothyroidism (35.3%, n=6). Grade 3-4 adverse events occurred in 47.1% of patients (n=8). There were 3 adverse events (17.6%) that led to discontinuation, of which 2 (11.8%) were grade 3-4. There were no grade 5 adverse events. Conclusion: Ipilimumab plus nivolumab in non-epithelial ovarian cancer resulted in an ORR of 12% (with 2 of 8 patients with granulosa ovarian tumors showing a durable CR and PR [both, >4 years)) and CBR of 29.4%, with durable responses seen. In contrast there were no responses in the carcinosarcoma group. One patient with Sertoli Leydig cell tumor suggested a possible benefit. Correlative studies to determine response and resistance markers are ongoing. Expanded prospective studies in granulosa tumor but not carcinosarcoma are warranted. Citation Format: Young Kwang Chae, Megan Othus, Sandip Pravin Patel, Kelly J. Wilkinson, Emily M. Whitman-Purves, Jayanthi Lea, John M. Schallenkamp, Nabil Adra, Leonard J. Appleman, Mitchell Alden, Jessica Thomes Pepin, John A. Ellerton, Andrew Poklepovic, Adam Walter, Murtuza M. Rampurwala, William R. Robinson, Liam Il-Young Chung, Christine M. McLeod, Helen X. Chen, Elad Sharon, Howard Streicher, Christopher W. Ryan, Charles D. Blanke, Razelle Kurzrock. A phase II basket trial of dual anti-CTLA-4 and anti-PD-1 blockade in rare tumors (DART) SWOG S1609: the non-epithelial ovarian tumor cohort [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 2 (Clinical Trials and Late-Breaking Research); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(8_Suppl):Abstract nr CT161.
- Published
- 2023
- Full Text
- View/download PDF
48. Abstract CT163: A phase II basket trial of dual anti-CTLA-4 and anti-PD-1 blockade in rare tumors (DART) SWOG S1609: the small cell carcinoma of the ovary, hypercalcemic type cohort
- Author
-
Young Kwang Chae, Megan Othus, Sandip P. Patel, Raid Aljumaily, Khine Z. Win, Tanya Pejovic, Sajeve S. Thomas, William R. Robinson, Liam Il-Young Chung, Christine M. McLeod, Helen X. Chen, Elad Sharon, Howard Streicher, Christopher W. Ryan, Charles D. Blanke, and Razelle Kurzrock
- Subjects
Cancer Research ,Oncology - Abstract
Background: Dual checkpoint inhibition with Anti-PD-1 and anti-CTLA4 checkpoint inhibitors have shown to be efficacious in many malignancies, but its potential role in various rare solid cancers is yet to be established. Small cell ovarian carcinoma, hypercalcemic type (SCCOHT) is a rare tumor characterized by loss of SMARC 2/4 function and so presents a novel paradigm for the treatment of SWI/SNF pathway deficient tumors (Petar Jelinic et al., 2018; Marc Tischkowitz et al., 2020). This study presents the first results of ipilimumab and nivolumab used in the SCCOHT cohort (#49) of the SWOG S1609 Dual Anti-CTLA-4 & Anti-PD-1 blockade in Rare Tumors (DART) trial. Methods: DART is a prospective, open-label, multicenter/multi-cohort phase 2 clinical trial of ipilimumab (1mg/kg intravenously every 6 weeks) plus nivolumab (240mg intravenously every 2 weeks). The primary endpoint includes objective response rate (ORR) (RECIST v1.1) (confirmed complete (CR) and partial responses (PR)). Secondary endpoints include progression-free survival (PFS), overall survival (OS), stable disease (SD) >6 months, and toxicity. Results: Five evaluable patients (median age 30) with SCCOHT were analyzed. Objective response rate was 20% (1 CR with 100% regression). The patient with CR has a duration of response (DoR) and OS of 35+ months. Another patient, showed unconfirmed PR with 81% regression (DoR 4 months), this patient went on to have confirmed iCR (CR confirmed by iRECIST) at around 24 months and has OS of 38+ months. At 12 months, 3 patients remain alive and 1 patient remains progression free; overall median PFS was 1.8 months (1.0-∞); median OS was 24 months (4.5-∞). The most common adverse events were fatigue, nausea, pruritus, dry mouth, maculo-papular rash and aspartate aminotransferase elevation (50%, n=2, respectively). There were two incidents (33.3%) of grade 3-4 adverse events. None of the adverse events led to discontinuation. There were no grade 5 adverse events. Conclusion: Ipilimumab plus nivolumab in five patients with the ultra-rare small cell ovarian carcinoma (hypercalcemic type) resulted in one CR durable at 35+ months and one unconfirmed PR with 81% regression. This is the first prospective study demonstrating efficacy of nivolumab and ipilimumab in this rare disease. Correlative studies to determine response and resistance markers are ongoing. Expanded prospective studies in small cell ovarian histologies are needed. Citation Format: Young Kwang Chae, Megan Othus, Sandip P. Patel, Raid Aljumaily, Khine Z. Win, Tanya Pejovic, Sajeve S. Thomas, William R. Robinson, Liam Il-Young Chung, Christine M. McLeod, Helen X. Chen, Elad Sharon, Howard Streicher, Christopher W. Ryan, Charles D. Blanke, Razelle Kurzrock. A phase II basket trial of dual anti-CTLA-4 and anti-PD-1 blockade in rare tumors (DART) SWOG S1609: the small cell carcinoma of the ovary, hypercalcemic type cohort [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 2 (Clinical Trials and Late-Breaking Research); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(8_Suppl):Abstract nr CT163.
- Published
- 2023
- Full Text
- View/download PDF
49. Abstract CT162: A phase II basket trial of dual anti-CTLA-4 and anti-PD-1 blockade in rare tumors (DART) SWOG S1609: the clear cell ovarian, endometrial, cervical cancer cohorts
- Author
-
Young Kwang Chae, Christopher W. Ryan, Naing Aung, William R. Robinson, Megan Othus, Elad Sharon, David M. O'Malley, Floortje J. Backes, Charles D. Blanke, Ramez N. Eskander, and Razelle Kurzrock
- Subjects
Cancer Research ,Oncology - Abstract
Background: Dual checkpoint inhibition with anti-PD-1 and anti-CTLA4 checkpoint inhibitors have proven to be efficacious in numerous malignancies. This study presents the first results of ipilimumab and nivolumab in the clear cell gynecologic cancer cohorts of the SWOG S1609 Dual Anti-CTLA-4 & Anti-PD-1 blockade in Rare Tumors (DART) trial. Methods: DART is a prospective, open-label, multicenter/multi-cohort phase 2 clinical trial of ipilimumab (1mg/kg IV every 6wk) plus nivolumab (240mg IV every 2wk). The primary endpoint was objective response rate (ORR) (RECIST v1.1) (confirmed CR and PR); progression-free survival (PFS), overall survival (OS), stable disease (SD) >6 months, and toxicity are secondary endpoints. Results: Evaluable patients were as follows: clear cell ovarian cancer (N=19); clear cell endometrial cancer (N=8); clear cell cervical (N=5) (median ages, 53, 66, and 59 years; cohorts 46, 45, and 42, respectively) In the clear cell ovarian cancer cohort, ORR was 21.1% [CR, 15.8%, n=3; PR, 5.3%, n=1]; clinical benefit rate (CBR) (includes stable disease ≥6 months) was 31.6% (6/19 patients). Among three patients with confirmed CR, two patients showed 100% regression (with ongoing response at 36+ months and 37+ months respectively), and the other patient showed 67% regression (due to lymph node < 1.0cm), but eventually progressed after 722 days. One confirmed PR patient achieved 75% regression (ongoing response at 53+ months). Of note, three patients achieved unconfirmed PR; one showed 34% regression (5 months); another, 38% (51.5+ months); and another, 58% regression (5 months). The ORR when including unconfirmed PR is 36.8% (7/19). Median PFS was 3.7 months (95% confidence interval (CI); 1.7-∞). Median OS was 21.7 months (6.4-∞). In the clear cell endometrial cancer cohort, ORR was 0%; CBR, 25% (2/8 patients). Of note, one patient achieved unconfirmed PR with 69% regression (4 months). The ORR when including unconfirmed PR is 12.5% (1/8). Median PFS was 2.0 months (95% confidence interval; 1.8-na). Median OS was 4.3 months (4.2-na). In the clear cell cervical cancer cohort, ORR was 0%; CBR, 20.0% (1/5 patients). Median PFS was 2.2 months (95% CI; 1.9-na). Median OS was 23.6 months (95% CI; 15.3-na). The most common adverse events, in the three cohorts combined, were nausea (37.5%, n=12), fatigue (34.4%, n=11), anorexia (31.2%, n=10), hypothyroidism (31.2%, n=10), and pruritus (28.1%, n=9). Grade 3-4 adverse events were reported in 17 cases (53.1%) with no grade 5 adverse events. Conclusion: Ipilimumab plus nivolumab in 19 clear cell ovarian cancer patients resulted in an ORR of 21.1% and CBR of 31.6%, with two durable CRs ongoing at 3+ years; CBR was 25% and 20%, respectively, in clear cell endometrial and cervical cohorts, with no objective responses, albeit with only 8 and 5 patients per cohort. Correlative studies to determine response/resistance markers are ongoing. Further prospective studies in rare gynecologic malignancies are warranted. Citation Format: Young Kwang Chae, Christopher W. Ryan, Naing Aung, William R. Robinson, Megan Othus, Elad Sharon, David M. O'Malley, Floortje J. Backes, Charles D. Blanke, Ramez N. Eskander, Razelle Kurzrock. A phase II basket trial of dual anti-CTLA-4 and anti-PD-1 blockade in rare tumors (DART) SWOG S1609: the clear cell ovarian, endometrial, cervical cancer cohorts [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 2 (Clinical Trials and Late-Breaking Research); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(8_Suppl):Abstract nr CT162.
- Published
- 2023
- Full Text
- View/download PDF
50. Abstract 6515: Pancreatic ductal adenocarcinoma (PDAC) early detection
- Author
-
Juan P. Hinestrosa, Harmeet Dhani, Gregor Schroeder, Jean M. Lewis, Heath I. Balcer, Razelle Kurzrock, Dove Keith, Rosalie Sears, and Paul Billings
- Subjects
Cancer Research ,Oncology - Abstract
Introduction Pancreatic ductal adenocarcinoma (PDAC) is the most common form of pancreatic cancer and is one of the deadliest cancer in the U.S., with a 5-year survival rate of just 11%. It is expected that ~62,000 new cases will be diagnosed in the US in 2023 and >70% of those will be at the regional or distant state, when curative resection is no longer an option. It is imperative that tools enabling early disease detection (stages I and II) are developed to improve patient survival outcomes. Here, we report the results of a study on the utility of extracellular vesicles (EVs) for early PDAC detection. Methods Our case-control training set was comprised of 75 pathologically confirmed PDAC cases (Stage I = 31, Stage II = 44) and 640 controls (including 11 subjects with pancreatitis) for EV biomarker and algorithm selection via machine learning. EVs from plasma were isolated using a proprietary technology, then EV-bound proteins of interest were analyzed via an immunoassay. A second independently collected cohort of 20 confirmed PDAC cases (Stage I = 10, Stage II = 10) and 27 controls (including 9 patients newly diagnosed with diabetes) was used for validation. Results The machine learning analysis using EV-isolated proteins generated a signature that utilized 8 different biomarkers, with an AUC of 0.989 (95% CI: 0.980 - 0.998) in the training set and an AUC of 0.987 (CI: 0.964 - 1.000) in the validation set. For the validation set, the sensitivity was 95% (CI: 76.4% - 99.1%) and the specificity was 92.6% (CI: 76.6% - 97.9%), with 10 out of 10 stage I cases and 9 out of 10 stage II cases correctly classified. Conclusions The ability to alter the future outcomes of PDAC patients will be predicated on development of a new generation of early-detection biomarkers and technologies. The use of EV protein signatures, a new class of cell-free biomarker, permits detection at high sensitivity and specificity, as demonstrated for the independently collected validation cohort. Studies aimed to assess this test for high-risk patients (family history of PDAC, known germline mutations, precursor lesions, hereditary pancreatitis, new onset diabetes will continue towards the establishment of real-world evidence. Citation Format: Juan P. Hinestrosa, Harmeet Dhani, Gregor Schroeder, Jean M. Lewis, Heath I. Balcer, Razelle Kurzrock, Dove Keith, Rosalie Sears, Paul Billings. Pancreatic ductal adenocarcinoma (PDAC) early detection. [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 1 (Regular and Invited Abstracts); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(7_Suppl):Abstract nr 6515.
- Published
- 2023
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.