22 results on '"Jardim, Denis L."'
Search Results
2. The Clinical and Molecular Profile of Lung Cancer Patients Harboring the TP53 R337H Germline Variant in a Brazilian Cancer Center: The Possible Mechanism of Carcinogenesis.
- Author
-
Lopes, Carlos D. H., Antonacio, Fernanda F., Moraes, Priscila M. G., Asprino, Paula F., Galante, Pedro A. F., Jardim, Denis L., de Macedo, Mariana P., Sandoval, Renata L., Katz, Artur, de Castro Jr., Gilberto, and Achatz, Maria Isabel
- Subjects
LUNG cancer ,CANCER patients ,LI-Fraumeni syndrome ,SYMPTOMS ,POLLUTION - Abstract
In southern and southeastern Brazil, the TP53 founder variant c.1010G>A (R337H) has been previously documented with a prevalence of 0.3% within the general population and linked to a heightened incidence of lung adenocarcinomas (LUADs). In the present investigation, we cover clinical and molecular characterizations of lung cancer patients from the Brazilian Li-Fraumeni Syndrome Study (BLISS) database. Among the 175 diagnosed malignant neoplasms, 28 (16%) were classified as LUADs, predominantly occurring in females (68%), aged above 50 years, and never-smokers (78.6%). Significantly, LUADs manifested as the initial clinical presentation of Li-Fraumeni Syndrome in 78.6% of cases. Molecular profiling was available for 20 patients, with 14 (70%) revealing EGFR family alterations. In total, 23 alterations in cancer driver genes were identified, comprising 7 actionable mutations and 4 linked to resistance against systemic treatments. In conclusion, the carriers of TP53 R337H demonstrate a predisposition to LUAD development. Furthermore, our results indicate that environmental pollution potentially impacts the carcinogenesis of lung tumors in the carriers of TP53 R337H. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
3. Low‐energy amplitude‐modulated electromagnetic field exposure: Feasibility study in patients with hepatocellular carcinoma.
- Author
-
Capareli, Fernanda, Costa, Frederico, Tuszynski, Jack A., Sousa, Micelange C., Setogute, Yone de C., Lima, Pablo D., Carvalho, Luciana, Santos, Elizabeth, Gumz, Brenda P., Sabbaga, Jorge, de Castria, Tiago B., Jardim, Denis L., Freitas, Daniela, Horvat, Natally, Bezerra, Regis O. F., Testagrossa, Leonardo, Costa, Tiago, Zanesco, Tatiana, Iemma, Antonio F., and Abou‐Alfa, Ghassan K.
- Subjects
ELECTROMAGNETIC fields ,FEASIBILITY studies ,QUALITY of life - Abstract
Background: Patients with advanced hepatocellular carcinoma (HCC) and poor liver function lack effective systemic therapies. Low‐energy electromagnetic fields (EMFs) can influence cell biological processes via non‐thermal effects and may represent a new treatment option. Methods: This single‐site feasibility trial enrolled patients with advanced HCC, Child‐Pugh A and B, Eastern Cooperative Oncology Group 0–2. Patients underwent 90‐min amplitude‐modulated EMF exposure procedures every 2–4 weeks, using the AutEMdev (Autem Therapeutics). Patients could also receive standard care. The primary endpoints were safety and the identification of hemodynamic variability patterns. Exploratory endpoints included health‐related quality of life (HRQoL), overall survival (OS). and objective response rate (ORR) using RECIST v1.1. Results: Sixty‐six patients with advanced HCC received 539 AutEMdev procedures (median follow‐up, 30 months). No serious adverse events occurred during procedures. Self‐limiting grade 1 somnolence occurred in 78.7% of patients. Hemodynamic variability during EMF exposure was associated with specific amplitude‐modulation frequencies. HRQoL was maintained or improved among patients remaining on treatment. Median OS was 11.3 months (95% confidence interval [CI]: 6.0, 16.6) overall (16.0 months [95% CI: 4.4, 27.6] and 12.0 months [6.4, 17.6] for combination therapy and monotherapy, respectively). ORR was 24.3% (32% and 17% for combination therapy and monotherapy, respectively). Conclusion: AutEMdev EMF exposure has an excellent safety profile in patients with advanced HCC. Hemodynamic alterations at personalized frequencies may represent a surrogate of anti‐tumor efficacy. NCT01686412. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
4. "Non-metastatic, Castration-resistant Prostate Cancer: Diagnostic and Treatment Recommendations by an Expert Panel from Brazil".
- Author
-
Jardim, Denis L., e Silva, Adriano Gonçalves, Lima Pompeo, Alexandre Saad Fere, Sarkis, Alvaro Sadek, Cardoso, Ana Paula Garcia, Sasse, Andre Deeke, Fay, Andre Poisl, Soares, Andrey, Pompeo, Antonio Carlos Lima, Carneiro, Arie, Kann, Ariel Galapo, Fogassa, Camilla, De Freitas Junior, Celso Heitor, Chade, Daher Cezar, Herchenhorn, Daniel, De Almeida, Daniel Vargas Pivato, Da Rosa, Diogo Augusto Rodrigues, Wiermann, Evanius Garcia, Schutz, Fabio Augusto Barros, and Kater, Fabio Roberto
- Subjects
CASTRATION-resistant prostate cancer ,ANTINEOPLASTIC agents ,PROSTATE-specific antigen ,ANDROGEN receptors ,FOLLOW-up studies (Medicine) - Abstract
A panel of Brazilian experts convened in order to provide recommendations regarding staging methods, antineoplastic therapy, osteoclast-targeted therapy, and patient follow-up in non-metastatic, castrationresistant prostate cancer. Key points include the reliance on prostate-specific antigen doubling time for treatment decisions, the absence of a clear preference between conventional and novel imaging techniques, and the increasing role of novel androgen-receptor signaling inhibitors. Introduction: Non-metastatic, castration-resistant prostate cancer (nmCRPC) is an important clinical stage of prostate cancer, prior to morbidity and mortality from clinical metastases. In particular, the introduction of novel androgenreceptor signaling inhibitors (ARSi) has changed the therapeutic landscape in nmCRPC. Given recent developments in this field, we update our recommendations for the management of nmCRPC. Methods: A panel of 51 invited medical oncologists and urologists convened in May of 2021 with the aim of discussing and providing recommendations regarding the most relevant issues concerning staging methods, antineoplastic therapy, osteoclast-targeted therapy, and patient follow-up in nmCRPC. Panel members considered the available evidence and their practical experience to address the 73 multiple-choice questions presented. Results: Key recommendations and findings include the reliance on prostate-specific antigen doubling time for treatment decisions, the absence of a clear preference between conventional and novel (i.e., positron-emission tomography-based) imaging techniques, the increasing role of ARSis in various settings, the general view that ARSis have similar efficacy. Panelists highlighted the slight preference for darolutamide, when safety is of greater concern, and a continued need to develop high-level evidence to guide the intensity of followup in this subset of prostate cancer. Discussion: Despite the limitations associated with a consensus panel, the topics addressed are relevant in current practice, and the recommendations can help practicing clinicians to provide state-of-the-art treatment to patients with nmCRPC in Brazil and other countries with similar healthcare settings. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
5. Comprehensive Landscape of Cyclin Pathway Gene Alterations and Co-occurrence with FGF/FGFR Aberrations Across Urinary Tract Tumors.
- Author
-
Jardim, Denis L F, Millis, Sherri Z, Ross, Jeffrey S, Lippman, Scott, Ali, Siraj M, and Kurzrock, Razelle
- Subjects
CANCER chemotherapy ,THERAPEUTIC use of antineoplastic agents ,FIBROBLAST growth factors ,GENETIC mutation ,CELL cycle proteins ,INDIVIDUALIZED medicine ,CELLULAR signal transduction ,URINARY organs ,TREATMENT effectiveness ,CELL cycle ,MOLECULAR biology ,CHROMOSOME abnormalities ,COMORBIDITY ,GENETIC profile - 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. Cyclin gene alterations are common in urinary tract tumors and often co-occur with FGF/FGFR aberrations. This article analyzes molecular alterations affecting the cyclin signals and potential co-drivers in key pathways. Rare subtypes of urinary tract cancers are included to identify unique alteration patterns and potential therapeutic opportunities. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
6. Interplay between Tumor Mutational Burden and Mutational Profile and Its Effect on Overall Survival: A Pilot Study of Metastatic Patients Treated with Immune Checkpoint Inhibitors.
- Author
-
Xavier, Camila B., Lopes, Carlos Diego H., Awni, Beatriz M., Campos, Eduardo F., Alves, João Pedro B., Camargo, Anamaria A., Guardia, Gabriela D. A., Galante, Pedro A. F., and Jardim, Denis L.
- Subjects
THERAPEUTIC use of monoclonal antibodies ,THERAPEUTIC use of antineoplastic agents ,PILOT projects ,RESEARCH ,STATISTICS ,IMMUNE checkpoint inhibitors ,GENETIC mutation ,PROGRAMMED death-ligand 1 ,MULTIVARIATE analysis ,AGE distribution ,CARCINOGENESIS ,METASTASIS ,TREATMENT effectiveness ,SEX distribution ,SURVIVAL analysis (Biometry) ,DESCRIPTIVE statistics ,TUMORS ,STATISTICAL correlation ,DATA analysis ,OVERALL survival ,LONGITUDINAL method ,PROPORTIONAL hazards models - Abstract
Simple Summary: Recently, patients with high-TMB tumors received agnostic FDA approval to be treated with pembrolizumab. However, some high-TMB patients do not show clinical benefits from this strategy. In this manuscript, we investigated a large cohort of 488 patients with TMB ≥ 10 mut/Mb treated with the following immune checkpoint inhibitors (ICIs), and correlated the clinical outcomes with the distinct somatic mutational profile of tumors: monoclonal antibody directed against programmed cell death protein-1 or monoclonal antibody directed against programmed cell death ligand 1 (anti-PD-1/anti-PD-L1); monoclonal antibody directed against cytotoxic T lymphocyte-associated antigen (anti-CTLA-4); combined treatment regimen including one anti-PD-1/anti-PD-L1 and one anti-CTLA-4 (ICIs combination). We know that some genomic alterations in TMB-high patients are already documented to help define prognosis and outcomes during immunotherapy. Conversely, other variables, such as MSI status, age, or gender, were not important to predict response to ICI treatment in this scenario, which could hypothesize the presence of a response prediction hierarchy. Thus, we believe that our manuscript is of broad interest to the general oncology community and can be used to better select patients for ICI treatment. Purpose: Solid tumors harboring tumor mutational burden (TMB) ≥10 mutations per megabase (mut/Mb) received agnostic approval for pembrolizumab. This work aims to analyze the somatic mutational profile's influence on the outcomes of patients with TMB-high tumors treated with immune checkpoint inhibitors (ICIs). Methods: This post-hoc analysis evaluated clinical and molecular features of patients with solid tumors treated with ICIs that could be either monoclonal antibody directed against programmed cell death protein-1 or monoclonal antibody directed against programmed cell death ligand 1 (anti-PD-1/anti-PD-L1), monoclonal antibody directed against cytotoxic T lymphocyte-associated antigen (anti-CTLA-4) or a combined treatment regimen including one anti-PD-1/anti-PD-L1 and one anti-CTLA-4 (ICIs combination). We performed OS analysis for TMB thresholds of ≥10, ≥20, and <10 mut/Mb. We assessed OS according to the mutational profile for a TMB ≥ 10 mut/Mb cutoff. For genes correlated with OS at the univariate assessment, we conducted a Cox multivariate analysis adjusted by median TMB, sex, age, microsatellite instability (MSI), and histology. Results: A total of 1661 patients were investigated; 488 with a TMB ≥10 mut/Mb (29.4%). The median OS was 42 months for TMB ≥10 or 20 mut/Mb, and 15 months for TMB <10 mut/Mb (p < 0.005). Among TMB ≥10 mut/Mb patients, mutations in E2F3 or STK11 correlated with worse OS, and mutations in NTRK3, PTPRD, RNF43, TENT5C, TET1, or ZFHX3 with better OS. These associations were confirmed with univariate and multivariate analyses (p < 0.05). Melanoma histology and TMB above the median endowed patients with better OS (p < 0.05), while MSI status, age, and gender did not have a statistically significant effect on OS. Conclusion: Combining TMB and mutation profiles in key cancer genes can better qualify patients for ICI treatment and predict their OS. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
7. Choice of 16S Ribosomal RNA Primers Impacts Male Urinary Microbiota Profiling.
- Author
-
Heidrich, Vitor, Inoue, Lilian T., Asprino, Paula F., Bettoni, Fabiana, Mariotti, Antonio C. H., Bastos, Diogo A., Jardim, Denis L. F., Arap, Marco A., and Camargo, Anamaria A.
- Subjects
RIBOSOMAL RNA ,HYPERVARIABLE regions ,DNA primers ,URINARY organs ,NUCLEOTIDE sequencing ,MICROBIAL communities - Abstract
Accessibility to next-generation sequencing (NGS) technologies has enabled the profiling of microbial communities living in distinct habitats. 16S ribosomal RNA (rRNA) gene sequencing is widely used for microbiota profiling with NGS technologies. Since most used NGS platforms generate short reads, sequencing the full-length 16S rRNA gene is impractical. Therefore, choosing which 16S rRNA hypervariable region to sequence is critical in microbiota profiling studies. All nine 16S rRNA hypervariable regions are taxonomically informative, but due to variability in profiling performance for specific clades, choosing the ideal 16S rRNA hypervariable region will depend on the bacterial composition of the habitat under study. Recently, NGS allowed the identification of microbes in the urinary tract, and urinary microbiota has become an active research area. However, there is no current study evaluating the performance of different 16S rRNA hypervariable regions for male urinary microbiota profiling. We collected urine samples from male volunteers and profiled their urinary microbiota by sequencing a panel of six amplicons encompassing all nine 16S rRNA hypervariable regions. Systematic comparisons of their performance indicate V1V2 hypervariable regions better assess the taxa commonly present in male urine samples, suggesting V1V2 amplicon sequencing is more suitable for male urinary microbiota profiling. We believe our results will be helpful to guide this crucial methodological choice in future male urinary microbiota studies. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
8. Landscape of Cyclin Pathway Genomic Alterations Across 5,356 Prostate Cancers: Implications for Targeted Therapeutics.
- Author
-
Jardim, Denis L., Millis, Sherri Z., Ross, Jeffrey S., Woo, Michelle Sue‐Ann, Ali, Siraj M., and Kurzrock, Razelle
- Subjects
PROSTATE tumors treatment ,SEQUENCE analysis ,CELL cycle proteins ,CELLULAR signal transduction ,GENOMICS ,GENE amplification - Abstract
The cyclin pathway may confer resistance to standard treatments but also offer novel therapeutic opportunities in prostate cancer. Herein, we analyzed prostate cancer samples (majority metastatic) using comprehensive genomic profiling performed by next‐generation sequencing (315 genes, >500× coverage) for alterations in activating and sensitizing cyclin genes (CDK4 amplification, CDK6 amplification, CCND1, CCND2, CCND3, CDKN2B [loss], CDKN2A [loss], SMARCB1), androgen receptor (AR) gene, and coalterations in genes leading to cyclin inhibitor therapeutic resistance (RB1 and CCNE1). Overall, cyclin sensitizing pathway genomic abnormalities were found in 9.7% of the 5,356 tumors. Frequent alterations included CCND1 amplification (4.2%) and CDKN2A and B loss (2.4% each). Alterations in possible resistance genes, RB1 and CCNE1, were detected in 9.7% (up to 54.6% in neuroendocrine) and 1.2% of cases, respectively, whereas AR alterations were seen in 20.9% of tumors (~27.3% in anaplastic). Cyclin sensitizing alterations were also more frequently associated with concomitant AR alterations. Clinical trials with cyclin inhibitors for prostate cancer are ongoing; thus, characterization of the landscape of cyclin pathway genomic alterations is needed. To that end, this article analyzes prostate cancer samples using comprehensive genomic profiling performed by next‐generation sequencing. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
9. Trametinib and Hydroxychloroquine (HCQ) Combination Treatment in KRAS-Mutated Advanced Pancreatic Adenocarcinoma: Detailed Description of Two Cases.
- Author
-
Xavier, Camila B., Marchetti, Katia R., Castria, Tiago B., Jardim, Denis L. F., and Fernandes, Gustavo S.
- Abstract
Purpose: Over the last decades, cytotoxic chemotherapy has been the cornerstone of metastatic pancreatic adenocarcinoma treatment. In late-stage disease, a range of treatment regimens still offers minor benefits. Molecular profiling studies have shown that pancreatic adenocarcinoma (PDAC) is a mutation-driven tumor type, with KRAS mutations found in approximately 90% of cases, which could partially explain the resistance to chemotherapy. Preclinical data on selective targeting of a downstream point of the RAF–MEK–ERK pathway with a MEK inhibitor along with the concurrent use of an autophagy inhibitor such as hydroxychloroquine appears to be one alternative approach to overcome resistance and inhibit cell proliferation. Methods: We herein aim to investigate the rationale of autophagy inhibitors use and describe the outcomes of patients who received this experimental treatment. Results: Two patients have received this experimental regimen from January 2020 to the present date, achieving disease stabilization that is clinically meaningful, considering the chemoresistance scenario of the included patients. Conclusions: Our real-life data regarding KRAS-mutated PDAC patients who received treatment with the MEK inhibitor trametinib combined with hydroxychloroquine after experiencing disease progression are consistent with the preclinical data, pointing to the clinical benefits of this regimen. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
10. Cyclin Pathway Genomic Alterations Across 190,247 Solid Tumors: Leveraging Large‐Scale Data to Inform Therapeutic Directions.
- Author
-
Jardim, Denis L., Millis, Sherri Z., Ross, Jeffrey S., Woo, Michelle Sue‐Ann, Ali, Siraj M., and Kurzrock, Razelle
- Subjects
TUMOR treatment ,GENOMICS - Abstract
Background: We describe the landscape of cyclin and interactive gene pathway alterations in 190,247 solid tumors. Methods: Using comprehensive genomic profiling (315 genes, >500× coverage), samples were analyzed for alterations in activating/sensitizing cyclin genes (CDK4 amplification, CDK6 amplification, CCND1, CCND2, CCND3, CDKN2B [loss], CDKN2A [loss], SMARCB1), hormone genes (estrogen receptor 1 [ESR1], androgen receptor [AR]), and co‐alterations in genes leading to cyclin inhibitor therapeutic resistance (RB1 and CCNE1). Results: Alterations in at least one cyclin activating/sensitizing gene occurred in 24% of malignancies. Tumors that frequently harbored at least one cyclin alteration were brain gliomas (47.1%), esophageal (40.3%) and bladder cancer (37.9%), and mesotheliomas (37.9%). The most frequent alterations included CDKN2A (13.9%) and CDKN2B loss (12.5%). Examples of unique patterns of alterations included CCND1 amplification in breast cancer (17.3%); CDK4 alterations in sarcomas (12%); CCND2 in testicular cancer (23.4%), and SMARCB1 mutations in kidney cancer (3% overall, 90% in malignant rhabdoid tumors). Alterations in resistance genes RB1 and CCNE1 affected 7.2% and 3.6% of samples. Co‐occurrence analysis demonstrated a lower likelihood of concomitant versus isolated alterations in cyclin activating/sensitizing and resistance genes (odds ratio [OR], 0.35; p <.001), except in colorectal, cervical, and small intestine cancers. AR and cyclin activating/sensitizing alterations in prostate cancer co‐occurred more frequently (vs. AR alterations and wild‐type cyclin activating/sensitizing alterations) (OR, 1.79; p <.001) as did ESR1 and cyclin activating/sensitizing alterations in breast (OR, 1.62; p <.001) and cervical cancer (OR, 4.08; p =.04) (vs. ESR1 and cyclin wild‐type activating/sensitizing alterations). Conclusion: Cyclin pathway alterations vary according to tumor type/histology, informing opportunities for targeted therapy, including for rare cancers. Implications for Practice: Cyclin pathway genomic abnormalities are frequent in human solid tumors, with substantial variation according to tumor site and histology. Opportunities for targeted therapy emerge with comprehensive profiling of this pathway. This article identifies molecular alterations in genes involved in the cyclin activation/sensitizing pathways and reports coexisting resistance and hormone pathway alterations in 190,247 diverse solid tumors that underwent next‐generation sequencing. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
11. Efficacy and safety of anticancer drug combinations: a meta-analysis of randomized trials with a focus on immunotherapeutics and gene-targeted compounds.
- Author
-
Jardim, Denis L., De Melo Gagliato, Débora, Nikanjam, Mina, Barkauskas, Donald A., and Kurzrock, Razelle
- Subjects
ANTINEOPLASTIC agents ,CLINICAL trials ,PROGRESSION-free survival ,SMALL molecules ,ODDS ratio - Abstract
Hundreds of trials are being conducted to evaluate combination of newer targeted drugs as well as immunotherapy. Our aim was to compare efficacy and safety of combination versus single non-cytotoxic anticancer agents. We searched PubMed (01/01/2001 to 03/06/2018) (and, for immunotherapy, ASCO and ESMO abstracts (2016 through March 2018)) for randomized clinical trials that compared a single non-cytotoxic agent (targeted, hormonal, or immunotherapy) versus a combination with another non-cytotoxic partner. Efficacy and safety endpoints were evaluated in a meta-analysis using a linear mixed-effects model (guidelines per PRISMA Report).We included 95 randomized comparisons (single vs. combination non-cytotoxic therapies) (59.4%, phase II; 41.6%, phase III trials) (29,175 patients (solid tumors)). Combinations most frequently included a hormonal agent and a targeted small molecule (23%). Compared to single non-cytotoxic agents, adding another non-cytotoxic drug increased response rate (odds ratio [OR]=1.61, 95%CI 1.40-1.84)and prolonged progression-free survival (hazard ratio [HR]=0.75, 95%CI 0.69-0.81)and overall survival (HR=0.87, 95%CI 0.81-0.94) (all p<0.001), which was most pronounced for the association between immunotherapy combinations and longer survival. Combinations also significantlyincreased the risk of high-grade toxicities (OR=2.42, 95%CI 1.98-2.97) (most notably for immunotherapy and small molecule inhibitors) and mortality at least possibly therapy related (OR: 1.33, 95%CI 1.15-1.53) (both p<0.001) (absolute mortality = 0.90% (single agent) versus 1.31% (combinations)) compared to single agents. In conclusion, combinations of non-cytotoxic drugs versus monotherapy in randomized cancer clinical trials attenuated safety, but increased efficacy, with the balance tilting in favor of combination therapy, based on the prolongation in survival. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
12. Phosphatidylinositol 3-kinase pathway genomic alterations in 60,991 diverse solid tumors informs targeted therapy opportunities.
- Author
-
Millis, Sherri Z., Jardim, Denis L., Albacker, Lee, Ross, Jeffrey S., Miller, Vincent A., Ali, Siraj M., and Kurzrock, Razelle
- Subjects
HEAD & neck cancer ,CARCINOSARCOMAS ,TUMORS ,MITOGEN-activated protein kinases ,CELL receptors ,CELLULAR signal transduction ,DRUG therapy ,COMPARATIVE studies ,GENES ,RESEARCH methodology ,MEDICAL cooperation ,PEPTIDES ,PHOSPHATASES ,PHOSPHOTRANSFERASES ,PROTEINS ,RESEARCH ,RESEARCH funding ,TRANSFERASES ,EVALUATION research ,ODDS ratio - Abstract
Background: The phosphatidylinositol 3-kinase (PI3K) pathway is frequently altered in cancer. This report describes the landscape of PI3K alterations in solid tumors as well as co-alterations serving as potential resistance/attenuation mechanisms.Methods: Consecutive samples were analyzed in a commercial Clinical Laboratory Improvement Amendment-certified laboratory using comprehensive genomic profiling performed by next-generation sequencing (315 genes). The co-alterations evaluated included the Erb-B2 receptor tyrosine kinase 2 (ERBB2), ERBB3, ERBB4, RAS, MET proto-oncogene tyrosine kinase (MET), and mitogen-activated protein kinase kinase (MAP2K) genes as well as tumor protein 53 (TP53), estrogen receptor 1 (ESR1), and androgen receptor (AR).Results: Alterations in any of 18 PI3K-pathway associated genes were identified in 44% of 60,991 tumors. Although single base and insertions/deletions (indels) were the most frequent alterations, copy number changes and rearrangements were identified in 11% and 0.9% of patients, respectively. Overall, the most frequently altered genes were PIK3 catalytic subunit α (PIK3CA) (13%), phosphatase and tensin homolog (PTEN) (9%), and serine/threonine kinase 11 (STK11) (5%). Tumor types that frequently harbored at least 1 PI3K alteration were uterine (77%), cervical (62%), anal (59%), and breast (58%) cancers. Alterations also were discerned frequently in tumors with carcinosarcoma (89%) and squamous cell carcinoma (62%) histologies. Tumors with a greater likelihood of co-occurring PI3K pathway and MAPK pathway alterations included colorectal cancers (odds ratio [OR], 1.64; P < .001), mesotheliomas (OR, 2.67; P = .024), anal cancers (OR, 1.98; P = .03), and nonsquamous head and neck cancers (OR, 2.03; P = .019). The co-occurrence of ESR1 and/or AR alterations with PI3K alterations was statistically significant in bladder, colorectal, uterine, prostate, and unknown primary cancers.Conclusions: Comprehensive genomic profiling reveals altered PI3K-related genes in 44% of solid malignancies, including rare disease and histology types. The frequency of alterations and the co-occurrence of resistance pathways vary by tumor type, directly affecting opportunities for targeted therapy. [ABSTRACT FROM AUTHOR]- Published
- 2019
- Full Text
- View/download PDF
13. Circulating Tumor DNA Detection in the Management of Anti-EGFR Therapy for Advanced Colorectal Cancer.
- Author
-
Knebel, Franciele H., Bettoni, Fabiana, da Fonseca, Leonardo G., Camargo, Anamaria A., Sabbaga, Jorge, and Jardim, Denis L.
- Subjects
CIRCULATING tumor DNA ,COLON cancer ,PSYCHOTHERAPY ,CANCER chemotherapy ,POLYMERASE chain reaction ,GENETIC mutation - Abstract
Background: Anti-EGFR antibodies are a standard care for advanced KRAS -wild type colorectal cancers. Circulating tumor DNA (ctDNA) monitoring during therapy can detect emergence of KRAS mutant clones and early resistance to therapy. Case Presentation: We describe a 61-years-old man presenting a metastatic and recurrent rectal cancer treated with different chemotherapy regimens. His tumor was KRAS wild-type based on tissue analysis and he was treated sequentially with cetuximab-based chemotherapy, chemotherapy alone and panitumumab-based chemotherapy. We performed sequential analysis of ctDNA using droplet digital PCR (ddPCR) and a commercial assay designed for the detection of frequent KRAS mutations during his clinical follow-up. Prior to the first cetuximab-based chemotherapy ctDNA analysis demonstrated an absence of KRAS mutations. Emergence of KRAS mutations in ctDNA occurred ~3 months after treatment initiation and preceded clinical and imaging progression in about 2 months. Fractional abundance of KRAS mutation rapidly increased to 70.7% immediately before a chemotherapy alone regimen was initiated. Interestingly, KRAS mutation abundance decreased significantly during the first two months of chemotherapy, reaching a fractional abundance of 3.0%, despite minimal clinical benefit with this therapy. Re-challenge with a different anti-EGFR antibody was attempted as later line, but high levels of KRAS mutations in ctDNA before therapy correlated with an absence of clinical benefit. Conclusions: The monitoring of resistance mutations in KRAS using ctDNA during the treatment of KRAS wild-type advanced colorectal cancers can detect the emergence of resistant clones prior to clinical progression. Dynamics of resistant clones may alter during periods on and off anti-EGFR antibodies, detecting window of opportunities for a re-challenge with these therapies. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
14. Lessons From the Development of the Immune Checkpoint Inhibitors in Oncology.
- Author
-
Jardim, Denis L., de Melo Gagliato, Débora, and Kurzrock, Razelle
- Abstract
Immunotherapies are becoming increasingly important in the treatment armamentarium of a variety of malignancies. Immune checkpoint inhibitors are the most representative drugs receiving regulatory approval over the past few years. In a recent study published in Clinical Cancer Research, we demonstrated that these agents are being developed faster than other prior anticancer therapies. All checkpoint inhibitors received priority review, being granted with at least one Food and Drug Administration expedited program. Hence, some of them are getting marketing approval after preliminary trials. The model continues to rely on phase I trials, designed with traditional models for dose definition, although a substantial number of patients are treated during the dose expansion cohorts. We demonstrated that efficacy and safety are reasonably predicted from the dose-finding portion of phase I trials with these agents, assuring a low treatment-related mortality for patients throughout the development process. In this article, we further discuss and summarize these findings and update some recent approval information for immune checkpoint inhibitors. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
15. Phase I study of the combination of crizotinib (as a MET inhibitor) and dasatinib (as a c-SRC inhibitor) in patients with advanced cancer.
- Author
-
Kato, Shumei, Jardim, Denis L., Johnson, Faye M., Subbiah, Vivek, Piha-Paul, Sarina, Tsimberidou, Apostolia M., Falchook, Gerald S., Karp, Daniel, Zinner, Ralph, Wheler, Jennifer, Janku, Filip, Fu, Siqing, Lim, JoAnn, Bean, Stacie, Nguyen, Ly, Urban, Susan, Browne, Elsa, Meric-Bernstam, Funda, and Hong, David S.
- Subjects
COMBINATION drug therapy ,CLINICAL trials ,DRUG toxicity ,PROSTATE tumors ,SARCOMA ,TUMORS ,PROTEIN-tyrosine kinase inhibitors ,TREATMENT effectiveness ,DASATINIB ,THERAPEUTICS - Abstract
Background Both MET and c-SRC are important mediators of cancer progression and there is cross talk between the two molecules. Preclinical studies have demonstrated combination of MET and c-SRC inhibitors is effective in multiple cancer types.Methods We analyzed the safety and efficacy of administering a c-SRC inhibitor (dasatinib) in combination with a MET inhibitor (crizotinib) in a two-arm concurrent phase I study. Arm A consisted of crizotinib fixed at 250 mg twice per day with escalation of dasatinib. Arm B consisted of dasatinib fixed at 140 mg daily with escalation of crizotinib. Endpoints included dose-limiting toxicities (DLTs), recommended phase II dose (RP2D), and response (RECIST 1.1).Results We enrolled 61 patients (arm A: 31, arm B: 30). The most common cancers were sarcoma (21%) and prostate cancer (16%). In Arm A, at dose level 2 (DL2), 40% (2/5) experienced DLTs. In the expanded DL1, 21% (4/19) experienced DLTs (all grade 3). In Arm B, at DL2, 50% (2/4) experienced DLTs. In the expanded DL1, 22% (4/18) experienced DLTs (all grade 3). RP2D was determined to be arm A, DL1 (250 mg crizotinib orally twice per day plus 50 mg dasatinib orally daily). Partial response (N = 1) and stable disease for ≥6 months (N = 3) were seen.Conclusions The combination of crizotinib and dasatinib is safe to administer but tolerability is limited given the high rate of adverse events. Responses and durable stable disease were limited. Further precision therapy approach using this specific combination may be difficult given the toxicity. [ABSTRACT FROM AUTHOR]- Published
- 2018
- Full Text
- View/download PDF
16. Impact of a Biomarker-Based Strategy on Oncology Drug Development: A Meta-analysis of Clinical Trials Leading to FDA Approval.
- Author
-
Fontes Jardim, Denis L., Schwaederle, Maria, Caimiao Wei, Lee, J. Jack, Hong, David S., Eggermont, Alexander M., Schilsky, Richard L., Mendelsohn, John, Lazar, Vladimir, Kurzrock, Razelle, Wei, Caimiao, and Jardim, Denis L
- Subjects
DRUG approval ,DRUG therapy ,CANCER treatment ,CANCER chemotherapy ,ANTINEOPLASTIC agents ,GOVERNMENT policy ,META-analysis ,RESEARCH funding ,TUMOR markers ,SYSTEMATIC reviews ,PHARMACODYNAMICS - Abstract
Background: In order to ascertain the impact of a biomarker-based (personalized) strategy, we compared outcomes between US Food and Drug Administration (FDA)-approved cancer treatments that were studied with and without such a selection rationale.Methods: Anticancer agents newly approved (September 1998 to June 2013) were identified at the Drugs@FDA website. Efficacy, treatment-related mortality, and hazard ratios (HRs) for time-to-event endpoints were analyzed and compared in registration trials for these agents. All statistical tests were two-sided.Results: Fifty-eight drugs were included (leading to 57 randomized [32% personalized] and 55 nonrandomized trials [47% personalized], n = 38 104 patients). Trials adopting a personalized strategy more often included targeted (100% vs 65%, P < .001), oral (68% vs 35%, P = .001), and single agents (89% vs 71%, P = .04) and more frequently permitted crossover to experimental treatment (67% vs 28%, P = .009). In randomized registration trials (using a random-effects meta-analysis), personalized therapy arms were associated with higher relative response rate ratios (RRRs, compared with their corresponding control arms) (RRRs = 3.82, 95% confidence interval [CI] = 2.51 to 5.82, vs RRRs = 2.08, 95% CI = 1.76 to 2.47, adjusted P = .03), longer PFS (hazard ratio [HR] = 0.41, 95% CI = 0.33 to 0.51, vs HR = 0.59, 95% CI = 0.53 to 0.65, adjusted P < .001) and a non-statistically significantly longer OS (HR = 0.71, 95% CI = 0.61 to 0.83, vs HR = 0.81, 95% CI = 0.77 to 0.85, adjusted P = .07) compared with nonpersonalized trials. Analysis of experimental arms in all 112 registration trials (randomized and nonrandomized) demonstrated that personalized therapy was associated with higher response rate (48%, 95% CI = 42% to 55%, vs 23%, 95% CI = 20% to 27%, P < .001) and longer PFS (median = 8.3, interquartile range [IQR] = 5 vs 5.5 months, IQR = 5, adjusted P = .002) and OS (median = 19.3, IQR = 17 vs 13.5 months, IQR = 8, Adjusted P = .04). A personalized strategy was an independent predictor of better RR, PFS, and OS, as demonstrated by multilinear regression analysis. Treatment-related mortality rate was similar for personalized and nonpersonalized trials.Conclusions: A biomarker-based approach was safe and associated with improved efficacy outcomes in FDA-approved anticancer agents. [ABSTRACT FROM AUTHOR]- Published
- 2015
- Full Text
- View/download PDF
17. Analysis of MET Genetic Aberrations in Patients With Breast Cancer at MD Anderson Phase I Unit.
- Author
-
Debora, de Melo Gagliato, Fontes Jardim, Denis L., Falchook, Gerald, Tang, Chad, Zinner, Ralph, Wheler, Jennifer J., Janku, Filip, Subbiah, Vivek, Piha-Paul, Sarina A., Fu, Siqing, Hess, Kenneth, Roy-Chowdhuri, Sinchita, Moulder, Stacy, Gonzalez-Angulo, Ana M., Meric-Bernstam, Funda, and Hong, David S.
- Published
- 2014
- Full Text
- View/download PDF
18. FBXW7 Mutations in Patients with Advanced Cancers: Clinical and Molecular Characteristics and Outcomes with mTOR Inhibitors.
- Author
-
Jardim, Denis L., Wheler, Jennifer J., Hess, Kenneth, Tsimberidou, Apostolia M., Zinner, Ralph, Janku, Filip, Subbiah, Vivek, Naing, Aung, Piha-Paul, Sarina A., Westin, Shannon N., Roy-Chowdhuri, Sinchita, Meric-Bernstam, Funda, and Hong, David S.
- Subjects
CANCER treatment ,GENETIC mutation ,MOLECULAR genetics ,HEALTH outcome assessment ,MTOR protein ,TUMOR suppressor genes - Abstract
Purpose: FBXW7 is a tumor suppressor gene responsible for the degradation of several proto-oncogenes. Preclinical data suggest that FBXW7 mutations sensitize cells to mTOR inhibitors. Clinicopathologic characteristics of cancer patients with FBXW7 mutations and their responses to mTOR inhibitors remain unknown. Methods: Using multiplex gene panels we evaluated how the FBXW7 mutation affected the cancer phenotype of patients referred to a phase I clinic starting January 2012. Whenever possible patients positive for FBXW7 mutation were treated with regimens containing an mTOR inhibitors and their outcomes were reviewed. Results: FBXW7 mutations were detected in 17 of 418 patients (4.0%). Among tumor types with more than 10 patients tested, FBXW7 mutations occurred in colorectal cancer (7/49; 14.3%), squamous cell cancer of head and neck (2/18; 11.1%), liver (1/13; 7.7%), and ovarian cancers (1/40; 2.5%). No one clinical, pathological or demographic feature was characteristic of the FBXW7-mutated patient population. The mutation occurred in isolation in only 2/17 (12%) patients, and KRAS was frequently found as a concomitant mutation, especially in patients with colorectal cancer (6/7; 86%). Ten patients were treated on a protocol containing an mTOR inhibitor, with a median time to treatment failure of 2.8 months (range, 1.3–6.8). One patient with liver cancer (fibrolamellar subtype) continues to have a prolonged stable disease for 6.8+ months. Conclusion: In patients with advanced cancers, somatic mutations in FBXW7 usually occur with other simultaneous molecular aberrations, which can contribute to limited therapeutic efficacy of mTOR inhibitors. [ABSTRACT FROM AUTHOR]
- Published
- 2014
- Full Text
- View/download PDF
19. Comprehensive characterization of malignant phyllodes tumor by whole genomic and proteomic analysis: biological implications for targeted therapy opportunities.
- Author
-
Jardim, Denis L. Fontes, Conley, Anthony, and Subbiah, Vivek
- Subjects
CANCER ,GENOMICS ,PROTEOMICS ,BIOLOGY ,GENETICS - Abstract
Background: Phyllodes tumors are uncommon breast tumors that account for less than 0.5% of all breast malignancies. After metastases develop, the prognosis is poor, with very few patients living more than 1 year. The biology of this unusual cancer is not understood and, consequently, no potential targets for treatments are currently available. There has been an exponential increase in the number of commercially available tumor profiling services. Herein, we report a case of metastatic malignant phyllodes tumor for which a comprehensive molecular analysis was performed by using Clinical Laboratory Improvement Amendments (CLIA)-certified labs, providing new insights into the potential opportunities for molecularly targeted therapies for this extremely rare disease.Methods: Next-generation sequencing was performed by using the FoundationOne™ platform (Foundation Medicine, Cambridge, MA). Whole-genome array-based comparative genomic hybridization (array CGH) was performed by using the DNAarray™ (CombiMatrix Diagnostics, Irvine, CA). Immunohistochemical and morphoproteomics analysis were performed at Consultative proteomics®, The University of Texas, UT Health Medical School, Houston,TX (Robert E Brown Lab); Clarient Diagnostics, Aliso Viejo, CA; and Caris Life Sciences Target one, Irving, TX, USA.Results: Next-generation sequencing showed 3 aberrant genes: activating mutation Q61L on NRAS; inactivating mutations Q504* and K740* on RB1; and TP53 loss. Whole-genome array-based comparative genomic hybridization (array CGH) revealed amplifications of chromosome (chr.) 1 (CKS1B gene), chr. 8 (MYC gene), and chr. 9 (CDKN2A gene) Deletions of chr. 17 (TP53), chr. 10 (GATA3), chr. 11 (FGF4 and CCND1 genes), and chr.22 (PDGFβ). Immunohistochemical analysis for relevant markers showed a positive staining for transducing-like enhancer of split (TLE) 3; secreted protein acidic and rich in cysteine (SPARC) was expressed at 2-3+ in the cytoplasm of the tumors cells, whereas mammalian target of rapamycin (mTOR) was expressed up to 2+ in the nuclei of the tumor cells.Conclusions: We describe for the first time an NRAS mutation with concomitant activation of PI3K/Akt/mTOR in phyllodes tumor. We also found markers for sensitivity to taxane-based therapies, especially albumin-bound paclitaxel. Exploring the biology of rare malignancies by CLIA certified labs may be reasonable strategy for the development of targeted treatments. [ABSTRACT FROM AUTHOR]- Published
- 2013
- Full Text
- View/download PDF
20. <italic>MET</italic> alterations detected in blood-derived circulating tumor DNA correlate with bone metastases and poor prognosis.
- Author
-
Ikeda, Sadakatsu, Schwaederle, Maria, Mohindra, Mandakini, Fontes Jardim, Denis L., and Kurzrock, Razelle
- Subjects
CANCER patients ,GASTROINTESTINAL cancer ,CIRCULATING tumor DNA ,BLOOD plasma ,BONE metastasis - Abstract
Background: We analyzed clinical associations of
MET alterations in the plasma of patients with diverse malignancies. Methods: Digital sequencing of circulating tumor DNA (ctDNA) (54–70 genes) was performed in 438 patients; 263 patients also had tissue sequencing (182–315 genes). The most represented tumor types were gastrointestinal (28.1%), brain (24.9%), and lung (23.2%). Most patients (71.2%) had recurrent/metastatic disease. Results:MET alterations were observed in 31 patients (7.1%) and correlated with bone metastasis (P = 0.007), withTP53 (P = 0.001) andPTEN (P = 0.003) abnormalities, and with an increased number of alterations (median, 4 vs 1,P = 0.001) (all multivariable analyses). Patients withMET alterations demonstrated a significantly shorter median time to metastasis/recurrence (1.0 vs 10.4 months,P = 0.044, multivariable) and a poorer survival (30.6 vs 58.4 months,P = 0.013, univariate only). Of the 31 patients withMET alterations, 18 also had tissue testing; only two also had tissueMET alterations (11.1%);MET alterations were detected at a lower frequency in tissue (1.14%) compared to ctDNA (7.1%), withP = 0.0002. Conclusions: In conclusion, the detection ofMET alterations by liquid biopsy is feasible.MET ctDNA alterations were associated with a poorer prognosis, higher numbers of genomic abnormalities, and bone metastases. The correlation with bone metastases may explain the higher frequency ofMET alterations in blood ctDNA than in tissue (since bones are rarely biopsied) and the previous observations of bone-predominant responses to MET inhibitors. The high number of co-altered genes suggests that MET inhibitors may need to be combined with other agents to induce/optimize responses. [ABSTRACT FROM AUTHOR]- Published
- 2018
- Full Text
- View/download PDF
21. MET in ovarian cancer.
- Author
-
Tang, Chad, Jardim, Denis L., and Hong, David
- Published
- 2014
- Full Text
- View/download PDF
22. Response.
- Author
-
Jardim, Denis L., Schwaederle, Maria, Lee, J. Jack, Caimiao Wei, Mendelsohn, John, Schilsky, Richard L., Kurzrock, Razelle, and Wei, Caimiao
- Abstract
A response from the author of the article "Impact of a Biomarker-Based Strategy on Oncology Drug Development: A Meta-analysis of Clinical Trials Leading to FDA Approval" in the previous issue is presented.
- Published
- 2016
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.