88 results on '"G. Batist"'
Search Results
2. Spatial distribution of the sport fishing in the state of Amazonas, northern Brazil
- Author
-
C. Lubich, F. Siqueira-Souza, G. Batista, and C. Freitas
- Subjects
fishing areas ,spatialization ,fisheries resources ,fisheries zoning ,Science ,Biology (General) ,QH301-705.5 ,Zoology ,QL1-991 ,Botany ,QK1-989 - Abstract
Abstract The objective of this study was to spatialize sport fishing operations and assess the frequency of the use of the fishing areas in the state of Amazonas by combining the Geographic Information System (GIS) approach and information available in the documents sent to the Instituto de Proteção Ambiental do Amazonas (IPAAM). Information on sport fishing tourism operations was gathered from the IPAAM database and fishing licenses (FLs). Data analysis was conducted utilizing descriptive analysis, and the spatialization of the locations was performed using Q-GIS software. From 2002 to August 2021, 163 requests for FLs were made. There was a decrease in the amount of first time FL requests, with a peak in 2018 and 2019, N=17 and N=18, respectively. The activity is conducted in 24 municipalities, with Barcelos (31.36%) and Presidente Figueiredo (17.75%) being the most popular. At the sub-basin level, sport fishing is notably present in the Negro, Amazonas, Aripuanã, Madeira, Purus and Solimões River basins. Overall, 26.38% of operations take place in conservation areas, specifically in sustainable development reserves (SDRs). Barcelos recorded the largest number of rivers used, with 15 rivers. These results can support future proposals for the sustainable management of fisheries through the zoning of fishing areas in the state of Amazonas.
- Published
- 2024
- Full Text
- View/download PDF
3. PCN252 TIME-TO-TREATMENT INITIATION (TTI) IN COMMUNITY INFUSION CLINICS: DECREASING WAIT TIMES FOR CANADIAN ONCOLOGY PATIENTS
- Author
-
A. Drinkwater, S. Anderson, P. Nijjar, C. Chiasson, A. Pabla, S.V. Sethi, S. Ghedira, A. Khan, J. Hopkins, G. Batist, B. Yap, and H. Wehbi
- Subjects
medicine.medical_specialty ,business.industry ,Health Policy ,Emergency medicine ,Public Health, Environmental and Occupational Health ,Time to treatment ,Medicine ,Oncology patients ,business - Published
- 2020
- Full Text
- View/download PDF
4. Combining Genome Surveillance and Metadata To Characterize the Diversity of Staphylococcus aureus Circulating in an Italian Hospital over a 9-Year Period
- Author
-
U. Postiglione, G. Batisti Biffignandi, M. Corbella, C. Merla, E. Olivieri, G. Petazzoni, E. J. Feil, C. Bandi, P. Cambieri, S. Gaiarsa, M. Brilli, and D. Sassera
- Subjects
Staphylococcus aureus ,antibiotic resistance ,community acquired ,genomic surveillance ,hospital acquired ,risk factors ,Microbiology ,QR1-502 - Abstract
ABSTRACT Staphylococcus aureus is an opportunistic pathogen and a leading cause of morbidity and mortality worldwide. Genomic-based surveillance has greatly improved our ability to track the emergence and spread of high-risk clones, but the full potential of genomic data is only reached when used in conjunction with detailed metadata. Here, we demonstrate the utility of an integrated approach by leveraging a curated collection of clinical and epidemiological metadata of S. aureus in the San Matteo Hospital (Italy) through a semisupervised clustering strategy. We sequenced 226 sepsis S. aureus samples, recovered over a period of 9 years. By using existing antibiotic profiling data, we selected strains that capture the full diversity of the population. Genome analysis revealed 49 sequence types, 16 of which are novel. Comparative genomic analyses of hospital- and community-acquired infection ruled out the existence of genomic features differentiating them, while evolutionary analyses of genes and traits of interest highlighted different dynamics of acquisition and loss between antibiotic resistance and virulence genes. Finally, highly resistant clones belonging to clonal complexes (CC) 8 and 22 were found to be responsible for abundant infections and deaths, while the highly virulent CC30 was responsible for rare but deadly episodes of infections. IMPORTANCE Genome sequencing is an important tool in clinical microbiology, as it allows in-depth characterization of isolates of interest and can propel genome-based surveillance studies. Such studies can benefit from ad hoc methods of sample selection to capture the genomic diversity present in a data set. Here, we present an approach based on clustering of antibiotic resistance profiles that allows optimal sample selection for bacterial genomic surveillance. We apply the method to a 9-year collection of Staphylococcus aureus from a large hospital in northern Italy. Our method allows us to sequence the genomes of a large variety of strains of this important pathogen, which we then leverage to characterize the epidemiology in the hospital and to perform evolutionary analyses on genes and traits of interest. These analyses highlight different dynamics of acquisition and loss between antibiotic resistance and virulence genes.
- Published
- 2023
- Full Text
- View/download PDF
5. Clinical proteomics reveals vulnerabilities in non-invasive breast ductal carcinoma and drives personalized treatment strategies.
- Author
-
Mitsa G, Florianova L, Lafleur J, Aguilar-Mahecha A, Zahedi RP, Del Rincon SV, Basik M, Borchers CH, and Batist G
- Abstract
Ductal carcinoma in situ (DCIS) is the most common type (80%) of non-invasive breast lesions in women. The lack of validated prognostic markers, limited patient numbers, and variable tissue quality have a significant impact on diagnosis, risk stratification, patient enrolment, and the results of clinical studies. Here, we performed label-free quantitative proteomics on 50 clinical formalin-fixed, paraffin embedded biopsies, validating 22 putative biomarkers from independent genetic studies. Our comprehensive proteomic phenotyping reveals more than 380 differentially expressed proteins and metabolic vulnerabilities, that can inform new therapeutic strategies for DCIS and invasive ductal carcinoma (IDC). Due to the readily druggable nature of proteins and metabolic enzymes or metabolism inhibitors, this study is of high interest for clinical research and pharmaceutical industry. To further evaluate our findings, and to promote the clinical translation of our study, we developed a highly multiplexed targeted proteomics assay for 90 proteins associated with cancer metabolism, RNA regulation and signature cancer pathways, such as PI3K/AKT/mTOR and EGFR/RAS/RAF.
- Published
- 2024
- Full Text
- View/download PDF
6. The Society for Immunotherapy of Cancer Perspective on Tissue-Based Technologies for Immuno-Oncology Biomarker Discovery and Application.
- Author
-
Monette A, Aguilar-Mahecha A, Altinmakas E, Angelos MG, Assad N, Batist G, Bommareddy PK, Bonilla DL, Borchers CH, Church SE, Ciliberto G, Cogdill AP, Fattore L, Hacohen N, Haris M, Lacasse V, Lie WR, Mehta A, Ruella M, Sater HA, Spatz A, Taouli B, Tarhoni I, Gonzalez-Kozlova E, Tirosh I, Wang X, and Gnjatic S
- Abstract
With immuno-oncology becoming the standard of care for a variety of cancers, identifying biomarkers that reliably classify patient response, resistance, or toxicity becomes the next critical barrier towards improving care. Multi-parametric, multi-omics, and computational platforms generating an unprecedented depth of data are poised to usher in the discovery of increasingly robust biomarkers for enhanced patient selection and personalized treatment approaches. Deciding which developing technologies to implement in clinical settings ultimately, applied either alone or in combination, relies on weighing pros and cons, from minimizing patient sampling to maximizing data outputs, and assessing reproducibility and representativeness of findings, while lessening data fragmentation towards harmonization. These factors are all assessed while taking into consideration the shortest turnaround time. The Society for Immunotherapy of Cancer (SITC) Biomarkers Committee convened to identify important advances in biomarker technologies and to address advances in biomarker discovery using multiplexed immunohistochemistry and immunofluorescence, their coupling to single cell transcriptomics, along with mass spectrometry-based quantitative and spatially resolved proteomics imaging technologies. We summarize key metrics obtained, ease of interpretation, limitations and dependencies, technical improvements, and outward comparisons of these technologies. By highlighting the most interesting recent data contributed by these technologies, and by providing ways to improve their outputs, we hope to guide correlative research directions and assist in their evolution towards becoming clinically useful in IO.
- Published
- 2024
- Full Text
- View/download PDF
7. Identification of a central network hub of key prognostic genes based on correlation between transcriptomics and survival in patients with metastatic solid tumors.
- Author
-
Lazar V, Raymond E, Magidi S, Bresson C, Wunder F, Berindan-Neagoe I, Tijeras-Rabaland A, Raynaud J, Onn A, Ducreux M, Batist G, Lassen U, Cilius Nielsen F, Schilsky RL, Rubin E, and Kurzrock R
- Abstract
Background: Dysregulated pathways in cancer may be hub addicted. Identifying these dysregulated networks for targeting might lead to novel therapeutic options., Objective: Considering the hypothesis that central hubs are associated with increased lethality, identifying key hub targets within central networks could lead to the development of novel drugs with improved efficacy in advanced metastatic solid tumors., Design: Exploring transcriptomic data (22,000 gene products) from the WINTHER trial ( N = 101 patients with various metastatic cancers), in which both tumor and normal organ-matched tissue were available., Methods: A retrospective in silico analysis of all genes in the transcriptome was conducted to identify genes different in expression between tumor and normal tissues (paired t -test) and to determine their association with survival outcomes using survival analysis (Cox proportional hazard regression algorithm). Based on the biological relevance of the identified genes, hub targets of interest within central networks were then pinpointed. Patients were grouped based on the expression level of these genes ( K -mean clustering), and the association of these groups with survival was examined (Cox proportional hazard regression algorithm, Forest plot, and Kaplan-Meier plot)., Results: We identified four key central hub genes- PLOD3, ARHGAP11A, RNF216 , and CDCA8 , for which high expression in tumor tissue compared to analogous normal tissue had the most significant correlation with worse outcomes. The correlation was independent of tumor or treatment type. The combination of the four genes showed the highest significance and correlation with the poorer outcome: overall survival (hazard ratio (95% confidence interval (CI)) = 10.5 (3.43-31.9) p = 9.12E-07 log-rank test in a Cox proportional hazard regression model). Findings were validated in independent cohorts., Conclusion: The expression of PLOD3, ARHGAP11A, RNF216 , and CDCA8 constitute, when combined, a prognostic tool, agnostic of tumor type and previous treatments. These genes represent potential targets for intercepting central hub networks in various cancers, offering avenues for novel therapeutic interventions., Competing Interests: V.L., C.B., and F.W. are full-time employees of the Worldwide Innovative Network (WIN) Association—WIN Consortium. WIN Association—WIN Consortium is the owner of the patent family entitled Digital Display. The inventors are V.L. and S.M. E.Ra. receives consulting and is a shareholder of SCOR Health Science, GenoScience Pharma, and Stromacare, is a shareholder of Axoltis, is a Scientific Director of Institut des Vaisseaux et du Sang (IVS), is a member of the Board of Directors of Institut National du Cancer (INCa France). S.M. receives consultancy from WIN Association—WIN Consortium. A.O. receives consulting fees from Roche Israel, MSD Israel, Boehringer Ingelheim, and AstraZeneca. M.D. reports consulting or advisory role: Amgen, AstraZeneca, Bayer, Daiichi Sankyo, GlaxoSmithKline, Ipsen, Merck Serono, MSD, Pierre Fabre, Roche, Servier, Sotio, and Zymeworks. Speakers’ bureau: Amgen, AstraZeneca, Bayer, Merck KgaA, MSD, Pierre Fabre, Pfizer, Roche, Servier, and Terumo. G.B. collaborates in formal clinical trial contracts, IITs, and in joint grants funded by Canadian and Quebec governments with Roche, Merck, Novartis, Astra-Zeneca, Bayer, Esperas, Aurka, Caprion, MRM. U.L. reports advisory board roles at Bayer, Pfizer, and Novartis. Research grants: BMS, Roche, Pfizer, Lilly, Incyte, and GSK. R.L.S. is the Chairman of the WIN Association—WIN Consortium. He declares Research Support: and continues to serve as the PI of the ASCO TAPUR trial. ASCO receives research grants from the following companies in support of this trial: AstraZeneca, Bayer, Boehringer-Ingelheim, Bristol Myers Squibb, Genentech, Lilly, Merck, Pfizer, Seagen, TAIHO. He does not personally receive any compensation from these companies; receives Consulting: he is a consultant to the following companies: Aadi Bioscience*Brylogyx, Cellworks*, Clarified Precision Medicine*, EQRx*, Illumina*, Scandion Oncology* ZephyrAI*, and receive compensation from those designated (*) in the last 2 years. Board Service: He is a member of the Board of Directors of the following companies and receives compensation from each: Clarified Precision Medicine and Leap Therapeutics. R.K. is Chief Medical Officer of the WIN Association—WIN Consortium. She has received research funding from Boehringer Ingelheim, Debiopharm, Foundation Medicine, Genentech, Grifols, Guardant, Incyte, Konica Minolta, MedImmune, Merck Serono, Omniseq, Pfizer, Sequenom, Takeda, and TopAlliance and the NCI; as well as consultant and/or speaker fees and/or advisory board/consultant for Actuate Therapeutics, AstraZeneca, Bicara Therapeutics, Inc., Biological Dynamics, Caris, Datar Cancer Genetics, Daiichi, EISAI, EOM Pharmaceuticals, Iylon, LabCorp, Merck, NeoGenomics, Neomed, Pfizer, Prosperdtx, Regeneron, Roche, TD2/Volastra, Turning Point Therapeutics, X-Biotech; has an equity interest in CureMatch Inc. and IDbyDNA; serves on the Board of CureMatch and CureMetrix, and is a co-founder of CureMatch. I.B.N., A.T.R., J.R., F.C.N., and E.Ru. report no competing interests. All authors have read the policy of conflicts of interests., (© The Author(s), 2024.)
- Published
- 2024
- Full Text
- View/download PDF
8. Feasibility and safety of endoscopic ultrasound-guided diffusing alpha emitter radiation therapy for advanced pancreatic cancer: Preliminary data.
- Author
-
Miller CS, Lecavalier-Barsoum M, Ma K, Santos Dutra M, Kaitoukov Y, Bahoric B, Tomic N, Dinelle F, Enger S, Batist G, Yang S, Laporta D, Kavan P, Sahai A, Roberge D, and Donath D
- Abstract
Background and study aims Pancreatic cancer is a devastating disease with limited locoregional treatment options. Diffusing alpha-emitter radiation therapy (Alpha DaRT), a novel cancer treatment using alpha-particle interstitial radiotherapy, may help address this challenge. The aim of this study was to evaluate the feasibility and safety of endoscopic ultrasound (EUS)-guided Alpha DaRT for advanced pancreatic cancer. Patients and methods Patients with inoperable locally advanced or metastatic pancreatic adenocarcinoma were treated with EUS-guided Alpha DaRT insertion. The Alpha DaRT sources were delivered into pancreatic tumors using a standard EUS needle with a novel proprietary applicator. Adverse events (AEs) were assessed based on the Common Terminology Criteria for Adverse Events version 5.0. Tumor response was evaluated by imaging 4 to 6 weeks post treatment. Results The first five patients were treated between March and September 2023. The procedure was technically successful in all cases, with Alpha DaRT sources inserted into the target tumor. Estimated gross tumor volume coverage ranged from 8% to 44%. Fourteen AEs were reported among three patients. Four were serious AEs, none of which was associated with the treatment, but rather, with disease progression or medical assistance in dying. Only two AEs (mild) were deemed possibly related to the study device. At the 35-day visit, two patients had progressive disease and three had stable disease, with one of the latter showing partial response 2 months post procedure. Conclusions Preliminary results from this first-in-human trial indicate that EUS-guided Alpha DaRT treatment for unresectable pancreatic cancer is feasible and safe, with no device-associated serious AEs. Further investigation of this promising novel modality is underway., Competing Interests: Conflict of Interest Corey S. Miller is a consultant for Alpha Tau Medical and Boston Scientific. Anand Sahai is a consultant for Boston Scientific. All other authors have no relevant conflicts., (The Author(s). This is an open access article published by Thieme under the terms of the Creative Commons Attribution-NonDerivative-NonCommercial-License, permitting copying and reproduction so long as the original work is given appropriate credit. Contents may not be used for commercial purposes, or adapted, remixed, transformed or built upon. (https://creativecommons.org/licenses/by-nc-nd/4.0/).)
- Published
- 2024
- Full Text
- View/download PDF
9. Real-World Outcomes of Incurable Cancer Patients Treated with Unlisted Anticancer Treatments in an Academic Center in Quebec, Canada.
- Author
-
Miller A, Panet F, Korsos V, Miller WH Jr, and Batist G
- Subjects
- Humans, Quebec, Female, Male, Antineoplastic Agents therapeutic use, Aged, Middle Aged, Academic Medical Centers, Treatment Outcome, Aged, 80 and over, Neoplasms drug therapy
- Abstract
Medical oncology is a rapidly evolving field, with new medications being discovered yearly, contributing to increased survival rates. However, accessing drugs in a timely manner can be challenging. In Quebec, Canada, a physician can prescribe an unlisted anticancer treatment through a regulated pathway under exceptional circumstances. We conducted a quality improvement study describing the outcomes of incurable cancer patients receiving unlisted anticancer therapy at the Jewish General Hospital between 2018 and 2019. Though our study did not include a comparator arm, unlisted anticancer therapies were associated with interesting median progression-free survival (11 months) and overall survival (25 months). Moreover, a large proportion of treatments, 44%, were subsequently reimbursed in the province of Quebec. Given the delay in anticancer drug reimbursement, this pathway is essential for timely access to oncology drugs. Such 'special access' programs will likely become increasingly important as precision medicine becomes the standard of practice.
- Published
- 2024
- Full Text
- View/download PDF
10. mTORC1-Driven Protein Translation Correlates with Clinical Benefit of Capivasertib within a Genetically Preselected Cohort of PIK3CA-Altered Tumors.
- Author
-
Sobsey CA, Froehlich BC, Mitsa G, Ibrahim S, Popp R, Zahedi RP, de Bruin EC, Borchers CH, and Batist G
- Subjects
- Humans, Female, Cell Line, Tumor, Mutation, Proteomics methods, Pyrroles pharmacology, Pyrroles therapeutic use, Proto-Oncogene Proteins c-akt metabolism, Proto-Oncogene Proteins c-akt genetics, Class I Phosphatidylinositol 3-Kinases genetics, Class I Phosphatidylinositol 3-Kinases metabolism, Mechanistic Target of Rapamycin Complex 1 metabolism, Mechanistic Target of Rapamycin Complex 1 genetics, Pyrimidines pharmacology, Pyrimidines therapeutic use, Protein Biosynthesis drug effects, Breast Neoplasms drug therapy, Breast Neoplasms genetics, Breast Neoplasms metabolism
- Abstract
Capivasertib is a potent selective inhibitor of AKT. It was recently FDA approved in combination with fulvestrant to treat HR+, HER2-negative breast cancers with certain genetic alteration(s) activating the PI3K pathway. In phase I trials, heavily pretreated patients with tumors selected for activating PI3K pathway mutations treated with capivasertib monotherapy demonstrated objective response rates of <30%. We investigated the proteomic profile associated with capivasertib response in genetically preselected patients and cancer cell lines. We analyzed samples from 16 PIK3CA-mutated patient tumors collected prior to capivasertib monotherapy in the phase I trial. PI3K pathway proteins were precisely quantified with immuno-Matrix-Assisted Laser Desorption/Ionization-mass spectrometry (iMALDI-MS). Global proteomic profiles were also obtained. Patients were classified according to response to capivasertib monotherapy: "clinical benefit (CB)" (≥12 weeks without progression, n = 7) or "no clinical benefit (NCB)" (progression in <12 weeks, n = 9). Proteins that differed between the patient groups were subsequently quantified in AKT1- or PIK3CA-altered breast cancer cell lines with varying capivasertib sensitivity. The measured concentrations of AKT1 and AKT2 varied among the PIK3CA-mutated tumors but did not differ between the CB and NCB groups. However, analysis of the global proteome data showed that translational activity was higher in tumors of the NCB vs. CB group. When reproducibly quantified by validated LC-MRM-MS assays, the same proteins of interest similarly distinguished between capivasertib-sensitive versus -resistant cell lines. The results provide further evidence that increased mTORC1-driven translation functions as a mechanism of resistance to capivasertib monotherapy. Protein concentrations may offer additional insights for patient selection for capivasertib, even among genetically preselected patients., Significance: Capivasertib's first-in-class FDA approval demonstrates its promise, yet there remains an opportunity to optimize its use. Our results provide new evidence that proteomics can stratify genetically preselected patients on clinical benefit. Characterization of the same profile in cell lines furnishes additional validation. Among PIK3CA-altered tumors, increased mTORC1-driven translation appears to confer intrinsic resistance. Assessing mTORC1 activation could therefore prove a useful complement to the existing genetic selection strategy for capivasertib., (©2024 The Authors; Published by the American Association for Cancer Research.)
- Published
- 2024
- Full Text
- View/download PDF
11. Brief Report: Impact of Anti-Cancer Treatments on Outcomes of COVID-19 in Patients With Thoracic Cancers: A CCC19 Registry Analysis.
- Author
-
Kulkarni AA, Hennessy C, Wilson G, Ramesh V, Hwang C, Awosika J, Bakouny Z, Khan H, Vilar-Compte D, McKay R, Jani C, Weissmann L, Griffiths E, Batist G, Bouganim N, Mavromatis B, Bashir B, Nguyen RH, Riess JW, Puc M, Kasi A, Berg S, Castillo DR, Hayes-Lattin B, Hosmer W, Flora D, Mishra S, French B, Warner JL, Lopes G, Peters S, and Florez N
- Subjects
- Humans, Male, Female, Aged, Middle Aged, Antineoplastic Agents therapeutic use, COVID-19 epidemiology, Registries, Thoracic Neoplasms drug therapy, SARS-CoV-2
- Abstract
Competing Interests: Disclosure The authors have stated that they have no conflicts of interest.
- Published
- 2024
- Full Text
- View/download PDF
12. SC912 inhibits AR-V7 activity in castration-resistant prostate cancer by targeting the androgen receptor N-terminal domain.
- Author
-
Yi Q, Han X, Yu HG, Chen HY, Qiu D, Su J, Lin R, Batist G, and Wu JH
- Subjects
- Male, Humans, Animals, Mice, Cell Line, Tumor, Cell Proliferation drug effects, Xenograft Model Antitumor Assays, Protein Domains, Apoptosis drug effects, Signal Transduction drug effects, Androgen Receptor Antagonists pharmacology, Androgen Receptor Antagonists therapeutic use, Prostatic Neoplasms, Castration-Resistant genetics, Prostatic Neoplasms, Castration-Resistant drug therapy, Prostatic Neoplasms, Castration-Resistant metabolism, Prostatic Neoplasms, Castration-Resistant pathology, Receptors, Androgen metabolism, Receptors, Androgen genetics
- Abstract
Androgen deprivation therapies (ADT) are the mainstay treatments for castration-resistant prostate cancer (CRPC). ADT suppresses the androgen receptor (AR) signaling by blocking androgen biosynthesis or inhibiting AR with antiandrogens that target AR's ligand-binding domain (LBD). However, the ADT's effect is short-lived, as the AR signaling inevitably arises again, which is frequently coupled with AR-V7 overexpression. AR-V7 is a truncated form of AR that lacks the LBD, thus being constitutively active in the absence of androgens and irresponsive to AR-LBD-targeting inhibitors. Though compelling evidence has tied AR-V7 to drug resistance in CRPC, pharmacological inhibition of AR-V7 is still an unmet need. Here, we discovered a small molecule, SC912, which binds to full-length AR as well as AR-V7 through AR N-terminal domain (AR-NTD). This pan-AR targeting relies on the amino acids 507-531 in the AR-NTD. SC912 also disrupted AR-V7 transcriptional activity, impaired AR-V7 nuclear localization and DNA binding. In the AR-V7 positive CRPC cells, SC912 suppressed proliferation, induced cell-cycle arrest, and apoptosis. In the AR-V7 expressing CRPC xenografts, SC912 attenuated tumor growth and antagonized intratumoral AR signaling. Together, these results suggested the therapeutic potential of SC912 for CRPC., (© 2024. The Author(s), under exclusive licence to Springer Nature Limited.)
- Published
- 2024
- Full Text
- View/download PDF
13. New Anticancer Drugs: Reliably Assessing "Value" While Addressing High Prices.
- Author
-
Stewart DJ, Bradford JP, Sehdev S, Ramsay T, Navani V, Rawson NSB, Jiang DM, Gotfrit J, Wheatley-Price P, Liu G, Kaplan A, Spadafora S, Goodman SG, Auer RAC, and Batist G
- Subjects
- Humans, Canada, Quality-Adjusted Life Years, Drug Costs, COVID-19, Neoplasms drug therapy, Neoplasms economics, SARS-CoV-2, Antineoplastic Agents therapeutic use, Antineoplastic Agents economics, Cost-Benefit Analysis methods
- Abstract
Countries face challenges in paying for new drugs. High prices are driven in part by exploding drug development costs, which, in turn, are driven by essential but excessive regulation. Burdensome regulation also delays drug development, and this can translate into thousands of life-years lost. We need system-wide reform that will enable less expensive, faster drug development. The speed with which COVID-19 vaccines and AIDS therapies were developed indicates this is possible if governments prioritize it. Countries also differ in how they value drugs, and generally, those willing to pay more have better, faster access. Canada is used as an example to illustrate how "incremental cost-effectiveness ratios" (ICERs) based on measures such as gains in "quality-adjusted life-years" (QALYs) may be used to determine a drug's value but are often problematic, imprecise assessments. Generally, ICER/QALY estimates inadequately consider the impact of patient crossover or long post-progression survival, therapy benefits in distinct subpopulations, positive impacts of the therapy on other healthcare or societal costs, how much governments willingly might pay for other things, etc. Furthermore, a QALY value should be higher for a lethal or uncommon disease than for a common, nonlethal disease. Compared to international comparators, Canada is particularly ineffective in initiating public funding for essential new medications. Addressing these disparities demands urgent reform.
- Published
- 2024
- Full Text
- View/download PDF
14. Progression-Free Survival Gain: A Reliable Primary End Point for Drug Registration That Can Accelerate Patient Access to Urgently Needed Therapies.
- Author
-
Stewart DJ, Ramsay T, Navani V, Liu G, Jiang DM, and Batist G
- Subjects
- Humans, Progression-Free Survival, Disease-Free Survival, Neoplasms drug therapy, Antineoplastic Agents therapeutic use
- Published
- 2024
- Full Text
- View/download PDF
15. The Clinical, Genomic, and Transcriptomic Landscape of BRAF Mutant Cancers.
- Author
-
Kazandjian S, Rousselle E, Dankner M, Cescon DW, Spreafico A, Ma K, Kavan P, Batist G, and Rose AAN
- Abstract
Background: BRAF mutations are classified into four molecularly distinct groups, and Class 1 (V600) mutant tumors are treated with targeted therapies. Effective treatment has not been established for Class 2/3 or BRAF Fusions. We investigated whether BRAF mutation class differed according to clinical, genomic, and transcriptomic variables in cancer patients., Methods: Using the AACR GENIE (v.12) cancer database, the distribution of BRAF mutation class in adult cancer patients was analyzed according to sex, age, primary race, and tumor type. Genomic alteration data and transcriptomic analysis was performed using The Cancer Genome Atlas., Results: BRAF mutations were identified in 9515 (6.2%) samples among 153,834, with melanoma (31%), CRC (20.7%), and NSCLC (13.9%) being the most frequent cancer types. Class 1 harbored co-mutations outside of the MAPK pathway (TERT, RFN43) vs. Class 2/3 mutations (RAS, NF1). Across all tumor types, Class 2/3 were enriched for alterations in genes involved in UV response and WNT/β-catenin. Pathway analysis revealed enrichment of WNT/β-catenin and Hedgehog signaling in non-V600 mutated CRC. Males had a higher proportion of Class 3 mutations vs. females (17.4% vs. 12.3% q = 0.003). Non-V600 mutations were generally more common in older patients (aged 60+) vs. younger (38% vs. 15% p < 0.0001), except in CRC (15% vs. 30% q = 0.0001). Black race was associated with non-V600 BRAF alterations (OR: 1.58; p < 0.0001)., Conclusions: Class 2/3 BRAFs are more present in Black male patients with co-mutations outside of the MAPK pathway, likely requiring additional oncogenic input for tumorigenesis. Improving access to NGS and trial enrollment will help the development of targeted therapies for non-V600 BRAF mutations.
- Published
- 2024
- Full Text
- View/download PDF
16. A Phase II Exploratory Study to Identify Biomarkers Predictive of Clinical Response to Regorafenib in Patients with Metastatic Colorectal Cancer Who Have Failed First-Line Therapy.
- Author
-
Gambaro K, Marques M, McNamara S, Couetoux du Tertre M, Hoffert C, Srivastava A, Schab A, Alcindor T, Langleben A, Sideris L, Abdelsalam M, Tehfe M, Couture F, Batist G, and Kavan P
- Subjects
- Humans, Biomarkers, Phenylurea Compounds therapeutic use, Proto-Oncogene Proteins p21(ras), Colonic Neoplasms, Pyridines
- Abstract
Single-agent regorafenib is approved in Canada for metastatic colorectal cancer (mCRC) patients who have failed previous lines of therapy. Identifying prognostic biomarkers is key to optimizing therapeutic strategies for these patients. In this clinical study (NCT01949194), we evaluated the safety and efficacy of single-agent regorafenib as a second-line therapy for mCRC patients who received it after failing first-line therapy with an oxaliplatin or irinotecan regimen with or without bevacizumab. Using various omics approaches, we also investigated putative biomarkers of response and resistance to regorafenib in metastatic lesions and blood samples in the same cohort. Overall, the safety profile of regorafenib seemed similar to the CORRECT trial, where regorafenib was administered as ≥ 2 lines of therapy. While the mutational landscape showed typical mutation rates for the top five driver genes (APC, KRAS, BRAF, PIK3CA, and TP53), KRAS mutations were enriched in intrinsically resistant lesions. Additional exploration of genomic-phenotype associations revealed several biomarker candidates linked to unfavorable prognoses in patients with mCRC using various approaches, including pathway analysis, cfDNA profiling, and copy number analysis. However, further research endeavors are necessary to validate the potential utility of these promising genes in understanding patients' responses to regorafenib treatment.
- Published
- 2023
- Full Text
- View/download PDF
17. Third-line treatment patterns in HER2-positive metastatic breast cancer: a retrospective analysis of real-world data in Canada.
- Author
-
Gambaro K, Groleau M, McNamara S, Awan A, Salem M, Abdelsalam M, St-Hilaire E, Vincent F, Carrier J, MacKay H, Provencher L, Boudreau D, Hamilou Z, Saad F, Ferrario C, Batist G, and Marques M
- Subjects
- Humans, Female, Lapatinib therapeutic use, Retrospective Studies, Receptor, ErbB-2 analysis, Receptor, ErbB-2 therapeutic use, Canada, Ado-Trastuzumab Emtansine therapeutic use, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Breast Neoplasms drug therapy
- Abstract
There is an increasing demand for real-world data pertaining to the usage of cancer treatments, especially in settings where no standard treatment is specifically recommended. This study presents the first real-world analysis of third-line treatment patterns in HER2-positive metastatic breast cancer (mBC) patients in Canada. The purpose was to assess evolution of clinical practice and identify unmet needs in post-second-line therapy. Retrospective data from medical records of 66 patients who received third-line treatment before 31st October 2018, and data from 56 patients who received third-line treatment after this date, extracted from the Personalize My Treatment (PMT) cancer patient registry, were analyzed. In the first cohort, the study revealed heterogeneity in the third-line setting, with trastuzumab, lapatinib, and T-DM1 being the main treatment options. Even though data were collected before the wide availability of tucatinib, neratinib and trastuzumab deruxtecan in Canada, the PMT cohort revealed the emergence of new therapeutic combinations and a shift from lapatinib usage to T-DM1 choice was observed. These findings underscore the evolving nature of third-line treatment strategies in Canada, a facet that is intrinsically tied to the availability of new drugs. The absence of a consensus on post-second-line treatment highlights the pressing need for more efficient therapeutic alternatives beyond the currently available options. This study not only offers valuable insights into the present landscape of third-line treatment in Canada but validates the significance and effectiveness of the PMT registry as a tool for generating pan-Canadian real-world evidence in oncology and its capacity to provide information on evolution of therapeutic practices., Competing Interests: Author MG was employed by company Knight Therapeutics Inc. This study received funding from Knight Therapeutics Inc. Knight Therapeutics had the following involvement with the study: CR cohort patient selection design and review of the manuscript., (Copyright © 2023 Gambaro, Groleau, McNamara, Awan, Salem, Abdelsalam, St-Hilaire, Vincent, Carrier, MacKay, Provencher, Boudreau, Hamilou, Saad, Ferrario, Batist and Marques.)
- Published
- 2023
- Full Text
- View/download PDF
18. Lasofoxifene versus fulvestrant for ER+/HER2- metastatic breast cancer with an ESR1 mutation: results from the randomized, phase II ELAINE 1 trial.
- Author
-
Goetz MP, Bagegni NA, Batist G, Brufsky A, Cristofanilli MA, Damodaran S, Daniel BR, Fleming GF, Gradishar WJ, Graff SL, Grosse Perdekamp MT, Hamilton E, Lavasani S, Moreno-Aspitia A, O'Connor T, Pluard TJ, Rugo HS, Sammons SL, Schwartzberg LS, Stover DG, Vidal GA, Wang G, Warner E, Yerushalmi R, Plourde PV, Portman DJ, and Gal-Yam EN
- Subjects
- Humans, Female, Fulvestrant adverse effects, Pyrrolidines therapeutic use, Aromatase Inhibitors, Mutation, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Receptor, ErbB-2 genetics, Receptor, ErbB-2 metabolism, Breast Neoplasms drug therapy, Breast Neoplasms genetics, Breast Neoplasms pathology
- Abstract
Background: Acquired estrogen receptor alpha (ER/ESR1) mutations commonly cause endocrine resistance in ER+ metastatic breast cancer (mBC). Lasofoxifene, a novel selective ER modulator, stabilizes an antagonist conformation of wild-type and ESR1-mutated ER-ligand binding domains, and has antitumor activity in ESR1-mutated xenografts., Patients and Methods: In this open-label, randomized, phase II, multicenter, ELAINE 1 study (NCT03781063), we randomized women with ESR1-mutated, ER+/human epidermal growth factor receptor 2 negative (HER2-) mBC that had progressed on an aromatase inhibitor (AI) plus a cyclin-dependent kinase 4/6 inhibitor (CDK4/6i) to oral lasofoxifene 5 mg daily or IM fulvestrant 500 mg (days 1, 15, and 29, and then every 4 weeks) until disease progression/toxicity. The primary endpoint was progression-free survival (PFS); secondary endpoints were safety/tolerability., Results: A total of 103 patients received lasofoxifene (n = 52) or fulvestrant (n = 51). The most current efficacy analysis showed that lasofoxifene did not significantly prolong median PFS compared with fulvestrant: 24.2 weeks (∼5.6 months) versus 16.2 weeks (∼3.7 months; P = 0.138); hazard ratio 0.699 (95% confidence interval 0.434-1.125). However, PFS and other clinical endpoints numerically favored lasofoxifene: clinical benefit rate (36.5% versus 21.6%; P = 0.117), objective response rate [13.2% (including a complete response in one lasofoxifene-treated patient) versus 2.9%; P = 0.124], and 6-month (53.4% versus 37.9%) and 12-month (30.7% versus 14.1%) PFS rates. Most common treatment-emergent adverse events with lasofoxifene were nausea, fatigue, arthralgia, and hot flushes. One death occurred in the fulvestrant arm. Circulating tumor DNA ESR1 mutant allele fraction (MAF) decreased from baseline to week 8 in 82.9% of evaluable lasofoxifene-treated versus 61.5% of fulvestrant-treated patients., Conclusions: Lasofoxifene demonstrated encouraging antitumor activity versus fulvestrant and was well tolerated in patients with ESR1-mutated, endocrine-resistant mBC following progression on AI plus CDK4/6i. Consistent with target engagement, lasofoxifene reduced ESR1 MAF, and to a greater extent than fulvestrant. Lasofoxifene may be a promising targeted treatment for patients with ESR1-mutated mBC and warrants further investigation., (Copyright © 2023 The Authors. Published by Elsevier Ltd.. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
19. A NRF2 inhibitor selectively sensitizes KEAP1 mutant tumor cells to cisplatin and gefitinib by restoring NRF2-inhibitory function of KEAP1 mutants.
- Author
-
Aboulkassim T, Tian X, Liu Q, Qiu D, Hancock M, Wu JH, and Batist G
- Subjects
- Humans, Gefitinib pharmacology, Cell Line, Tumor, Kelch-Like ECH-Associated Protein 1 genetics, Kelch-Like ECH-Associated Protein 1 metabolism, Signal Transduction, Cisplatin pharmacology, NF-E2-Related Factor 2 metabolism
- Abstract
NRF2 (nuclear factor erythroid 2-related factor 2) is a master regulator of protective responses in healthy tissues. However, when it is active in tumor cells, it can result in drug resistance. KEAP1, the endogenous NRF2 inhibitor, binds NRF2 and redirects it to proteasomal degradation, so the KEAP1/NRF2 interaction is critical for maintaining NRF2 at a basal level. A number of clinically relevant KEAP1 mutations were shown to disrupt this critical KEAP1/NRF2 interaction, leading to elevated NRF2 levels and drug resistance. Here, we describe a small-molecule NRF2 inhibitor, R16, that selectively binds KEAP1 mutants and restores their NRF2-inhibitory function by repairing the disrupted KEAP1/NRF2 interactions. R16 substantially sensitizes KEAP1-mutated tumor cells to cisplatin and gefitinib, but does not do so for wild-type KEAP1 cells, and sensitizes KEAP1 G333C-mutated xenograft to cisplatin. We developed a BRET
2 -based biosensor system to detect the KEAP1/NRF2 interaction and classify KEAP1 mutations. This strategy would identify drug-resistant KEAP1 somatic mutations in clinical molecular profiling of tumors., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2023 The Authors. Published by Elsevier Inc. All rights reserved.)- Published
- 2023
- Full Text
- View/download PDF
20. A transcriptomics approach to expand therapeutic options and optimize clinical trials in oncology.
- Author
-
Lazar V, Zhang B, Magidi S, Le Tourneau C, Raymond E, Ducreux M, Bresson C, Raynaud J, Wunder F, Onn A, Felip E, Tabernero J, Batist G, Kurzrock R, Rubin E, and Schilsky RL
- Abstract
Background: The current model of clinical drug development in oncology displays major limitations due to a high attrition rate in patient enrollment in early phase trials and a high failure rate of drugs in phase III studies., Objective: Integrating transcriptomics for selection of patients has the potential to achieve enhanced speed and efficacy of precision oncology trials for any targeted therapies or immunotherapies., Methods: Relative gene expression level in the metastasis and normal organ-matched tissues from the WINTHER database was used to estimate in silico the potential clinical benefit of specific treatments in a variety of metastatic solid tumors., Results: As example, high mRNA expression in tumor tissue compared to analogous normal tissue of c-MET and its ligand HGF correlated in silico with shorter overall survival (OS; p < 0.0001) and may constitute an independent prognostic marker for outcome of patients with metastatic solid tumors, suggesting a strategy to identify patients most likely to benefit from MET-targeted treatments. The prognostic value of gene expression of several immune therapy targets (PD-L1, CTLA4, TIM3, TIGIT, LAG3, TLR4) was investigated in non-small-cell lung cancers and colorectal cancers (CRCs) and may be useful to optimize the development of their inhibitors, and opening new avenues such as use of anti-TLR4 in treatment of patients with metastatic CRC., Conclusion: This in silico approach is expected to dramatically decrease the attrition of patient enrollment and to simultaneously increase the speed and detection of early signs of efficacy. The model may significantly contribute to lower toxicities. Altogether, our model aims to overcome the limits of current approaches., Competing Interests: − Dr. Vladimir Lazar, Catherine Bresson, and Fanny Wunder are full-time employees of Worldwide Innovative Network (WIN) Association – WIN Consortium. − Shai Magidi receives consultancy fees from Worldwide Innovative Network (WIN) Association – WIN Consortium. − Worldwide Innovative Network (WIN) Association – WIN Consortium is the owner of the patent family entitled Digital Display. The inventors are Dr. Vladimir Lazar and Shai Magidi. − Dr. Baolin Zhang disclaimer: The views expressed in this manuscript are those of the author and do not represent the views or policies of the U.S. FDA. − Pr. Christophe Le Tourneau has participated in advisory boards from Celgene, AstraZeneca, MSD, BMS, Merck Serono, Nanobiotix, Rakuten, Seatlle Genetics, and Roche. − Pr. Eric Raymond : SCOR Health Science – consulting and shareholder; GenoScience Pharma – consulting and shareholder, Axoltis – shareholder, Stromacare – consulting and shareholderInstitut des Vaisseaux et du Sang (IVS) – Scientific Director, Institut National du Cancer (INCa France) – Member of the Board of Directors. − Pr. Michel Ducreux reports consulting or advisory role: Amgen, AstraZeneca, Bayer, Daiichi Sankyo, Merck Serono, MSD, Pierre Fabre, Roche, Servier. Speakers’ bureau: AstraZeneca, Bayer, Roche, Terumo Amir Onn receives consulting fees from: Roche Israel, MSD Israel, Boehringer Ingelheim, and AstraZeneca. − Enriqueta Felip reports consulting or advisory role: Abbvie, Amgen, AstraZeneca, Bayer, Blue Print Medicines, Boehringer Ingelheim, Bristol-Myers Squibb, Eli Lilly, GSK, Janssen, Merck, MSD, Novartis, Pfizer, Puma Biotechnology, Roche, Sanofi Genzyme, Takeda; Speakers’ bureau or expert testimony: AstraZeneca, Boehringer Ingelheim, Bristol-Myers Squibb, Eli Lilly, Medscape, MSD, Novartis, Peervoice, Pfizer, Prime Oncology, Roche, Springer, Takeda, Touchime, CME Outfitters; Research grant or funding: Grant for Oncology Innovation (GOI), Fundación Merck Salud; Other: Grífols (independent member of the board). − Dr. Josep Tabernero declares scientific consultancy role for Array Biopharma, AstraZeneca, Bayer, BeiGene, Boehringer Ingelheim, Chugai, Genentech, Inc., Genmab A/S, Halozyme, Imugene Limited, Inflection Biosciences Limited, Ipsen, Kura Oncology, Lilly, MSD, Menarini, Merck Serono, Merrimack, Merus, Molecular Partners, Novartis, Peptomyc, Pfizer, Pharmacyclics, ProteoDesign SL, Rafael Pharmaceuticals, F. Hoffmann-La Roche Ltd, Sanofi, SeaGen, Seattle Genetics, Servier, Symphogen, Taiho, VCN Biosciences, Biocartis, Foundation Medicine, HalioDX SAS and Roche Diagnostics. − Gerald Batist collaborates in formal clinical trial contracts, IITs and in joint grants funded by Canadian and Quebec governments with Roche, Merck, Novartis, AstraZeneca, Bayer, Esperas, Aurka, Caprion, MRM. − Razelle Kurzrock is Chief Medical Officer of the Worldwide Innovative Network (WIN) Association – WIN Consortium. She has received research funding from Genentech, Merck Serono, Pfizer, Boehringer Ingelheim, TopAlliance, Takeda, Incyte, Debiopharm, Medimmune, Sequenom, Foundation Medicine, Konica Minolta, Grifols, Omniseq, and Guardant, as well as consultant and/or speaker fees and/or advisory board for X-Biotech, Neomed, Pfizer, Actuate Therapeutics, Roche, Turning Point Therapeutics, TD2/Volastra, Bicara Therapeutics, Inc., has an equity interest in IDbyDNA and CureMatch Inc, serves on the Board of CureMatch and CureMetrix, and is a co-founder of CureMatch. − Richard L. Schilsky is the Chairman of Worldwide Innovative Network (WIN) Association – WIN Consortium. He declares Research Support: continues to serve as the PI of the ASCO TAPUR trial. ASCO receives research grants from the following companies in support of this trial: AstraZeneca, Bayer, Boehringer-Ingelheim, Bristol Myers Squibb, Genentech, Lilly, Merck, Pfizer, Seagen. He does not personally receive any compensation from these companies; receives Consulting: he is consultant to the following companies: Brylogyx, Cellworks*, Clarified Precision Medicine*, EQRx*, Illumina*, Scandion Oncology* and receive compensation from those designated (*). − Dr. Baolin Zhang, Dr. Jacques Raynaud and Pr. Eitan Rubin, declare no potential conflicts of interest., (© The Author(s), 2023.)
- Published
- 2023
- Full Text
- View/download PDF
21. Comorbidity between lung cancer and COVID-19 pneumonia: role of immunoregulatory gene transcripts in high ACE2 -expressing normal lung.
- Author
-
Lazar V, Raynaud J, Magidi S, Bresson C, Martini JF, Galbraith S, Wunder F, Onn A, Batist G, Girard N, Lassen U, Pramesh CS, Al-Omari A, Ikeda S, Berchem G, Blay JY, Solomon B, Felip E, Tabernero J, Rubin E, Philip T, Porgador A, Berindan-Neagoe I, Schilsky RL, and Kurzrock R
- Abstract
Background: SARS-CoV-2 (COVID-19) elicits a T-cell antigen-mediated immune response of variable efficacy. To understand this variability, we explored transcriptomic expression of angiotensin-converting enzyme 2 ( ACE2 , the SARS-CoV-2 receptor) and of immunoregulatory genes in normal lung tissues from patients with non-small cell lung cancer (NSCLC)., Methods: This study used the transcriptomic and the clinical data for NSCLC patients generated during the CHEMORES study [ n = 123 primary resected (early-stage) NSCLC] and the WINTHER clinical trial ( n = 32 metastatic NSCLC)., Results: We identified patient subgroups with high and low ACE2 expression ( p = 1.55 × 10
-19 ) in normal lung tissue, presumed to be at higher and lower risk, respectively, of developing severe COVID-19 should they become infected. ACE2 transcript expression in normal lung tissues (but not in tumor tissue) of patients with NSCLC was higher in individuals with more advanced disease. High- ACE2 expressors had significantly higher levels of CD8+ cytotoxic T lymphocytes and natural killer cells but with presumably impaired function by high Thymocyte Selection-Associated High Mobility Group Box Protein TOX ( TOX ) expression. In addition, immune checkpoint-related molecules - PD-L1, CTLA-4, PD-1 , and TIGIT - are more highly expressed in normal (but not tumor) lung tissues; these molecules might dampen immune response to either viruses or cancer. Importantly, however, high inducible T-cell co-stimulator ( ICOS ), which can amplify immune and cytokine reactivity, significantly correlated with high ACE2 expression in univariable analysis of normal lung (but not lung tumor tissue)., Conclusions: We report a normal lung immune-tolerant state that may explain a potential comorbidity risk between two diseases - NSCLC and susceptibility to COVID-19 pneumonia. Further, a NSCLC patient subgroup has normal lung tissue expressing high ACE2 and high ICOS transcripts, the latter potentially promoting a hyperimmune response, and possibly leading to severe COVID-19 pulmonary compromise., (© The Author(s), 2022.)- Published
- 2022
- Full Text
- View/download PDF
22. Transcriptomics in Tumor and Normal Lung Tissues Identify Patients With Early-Stage Non-Small-Cell Lung Cancer With High Risk of Postsurgery Recurrence Who May Benefit From Adjuvant Therapies.
- Author
-
Lazar V, Girard N, Raymond E, Martini JF, Galbraith S, Raynaud J, Bresson C, Solomon B, Magidi S, Nechushtan H, Onn A, Berger R, Chen H, Al-Omari A, Ikeda S, Lassen U, Sekacheva M, Felip E, Tabernero J, Batist G, Spatz A, Pramesh CS, Girard P, Blay JY, Philip T, Berindan-Neagoe I, Porgador A, Rubin E, Kurzrock R, and Schilsky RL
- Subjects
- B7-H1 Antigen analysis, CTLA-4 Antigen genetics, Humans, Lung chemistry, Prospective Studies, Retrospective Studies, Transcriptome, Carcinoma, Non-Small-Cell Lung genetics, Lung Neoplasms drug therapy, Small Cell Lung Carcinoma
- Abstract
Purpose: The prognosis of patients with non-small-cell lung cancer (NSCLC), traditionally determined by anatomic histology and TNM staging, neglects the biological features of the tumor that may be important in determining patient outcome and guiding therapeutic interventions. Identifying patients with NSCLC at increased risk of recurrence after curative-intent surgery remains an important unmet need so that known effective adjuvant treatments can be offered to those at highest risk of recurrence., Methods: Relative gene expression level in the primary tumor and normal bronchial tissues was used to retrospectively assess their association with disease-free survival (DFS) in a cohort of 120 patients with NSCLC who underwent curative-intent surgery., Results: Low versus high Digital Display Precision Predictor (DDPP) score (a measure of relative gene expression) was significantly associated with shorter DFS (highest recurrence risk; P = .006) in all patients and in patients with TNM stages 1-2 ( P = .00051; n = 83). For patients with stages 1-2 and low DDPP score (n = 29), adjuvant chemotherapy was associated with improved DFS ( P = .0041). High co-overexpression of CTLA-4 , PD-L1 , and ICOS in normal lung (28 of 120 patients) was also significantly associated with decreased DFS ( P = .0013), suggesting an immune tolerance to tumor neoantigens in some patients. Patients with DDPP low and immunotolerant normal tissue had the shortest DFS ( P = 2.12E-11)., Conclusion: TNM stage, DDPP score, and immune competence status of normal lung are independent prognostic factors in multivariate analysis. Our findings open new avenues for prospective prognostic assessment and treatment assignment on the basis of transcriptomic profiling of tumor and normal lung tissue in patients with NSCLC., Competing Interests: Vladimir LazarPatents, Royalties, Other Intellectual Property: I am inventor on patentes filed by WIN Consortium I do not receive money Nicolas GirardEmployment: AstraZeneca (I)Consulting or Advisory Role: Roche, Lilly, AstraZeneca, Novartis, Pfizer, Bristol Myers Squibb, MSD, Takeda, Janssen, Sanofi, AmgenResearch Funding: Roche (Inst), AstraZeneca (Inst), BMS (Inst)Travel, Accommodations, Expenses: Roche Eric RaymondEmployment: Genoscience Pharma, SCOR, StromaCare, Onward TherapeuticsLeadership: AFR-OncologyStock and Other Ownership Interests: Neuronax, Oncoethix, Genoscience Pharma, SCORConsulting or Advisory Role: Lilly, Pfizer, Merck Serono, PharmaEngine, Novartis/lpsen, Celgene Jean-François MartiniEmployment: PfizerStock and Other Ownership Interests: Pfizer Susan GalbraithEmployment: AstraZenecaLeadership: BB Biotech VenturesStock and Other Ownership Interests: AstraZeneca Benjamin SolomonConsulting or Advisory Role: BayerResearch Funding: AstraZeneca/Merck (Inst)Open Payments Link: https://openpaymentsdata.cms.gov/physician/358846 Hovav NechushtanResearch Funding: AstraZeneca (Inst), Spectrum Pharmaceuticals (Inst), Lilly (Inst), Merck KGaA (Inst)Uncompensated Relationships: MSD Amir OnnHonoraria: Boehringer Ingelheim, Roche, MSD, AmgenConsulting or Advisory Role: Boehringer Ingelheim, MSDSpeakers' Bureau: AstraZeneca Raanan BergerStock and Other Ownership Interests: BelongHonoraria: Bristol Myers Squibb, Roche Israel, AstraZeneca, Pfizer, MSDConsulting or Advisory Role: Belong, MSD Oncology, BMSSpeakers' Bureau: Mitra Biotech, BMS, MSD OncologyTravel, Accommodations, Expenses: Mitra Biotech, Bristol Myers Squibb, MSD, AstraZeneca Sadakatsu IkedaHonoraria: Chugai Pharma, Novartis, AstraZeneca, Taiho Pharmaceutical, Guardant Health, Bristol Myers Squibb-Ono Pharmaceutical, MSD, ACT Genomics, Roche, Canon Medical System, ACT MedConsulting or Advisory Role: Genodive Pharma (Inst)Research Funding: ACT Genomics (Inst), Hitachi (Inst), Canon Medical System (Inst) Ulrik LassenHonoraria: Bayer, Pfizer, NovartisConsulting or Advisory Role: Bayer, PfizerResearch Funding: BMS (Inst), Roche (Inst), Pfizer (Inst), GlaxoSmithKline (Inst), Incyte (Inst), Lilly (Inst) Enriqueta FelipConsulting or Advisory Role: Amgen, AstraZeneca, Bayer, Bristol Myers Squibb, Lilly, GlaxoSmithKline, Janssen, Merck Serono, Novartis, Pfizer, Sanofi, Takeda, Peptomyc, Daiichi Sankyo Europe GmbH, F. Hoffmann La Roche, Merck Sharp & DohmeSpeakers’ Bureau: AstraZeneca, Bristol Myers Squibb, Lilly, Medscape, Merck Sharp & Dohme, PeerVoice, Pfizer, Takeda, Amgen, F. Hoffmann La Roche, Janssen, Medical Trends, Merck Serono, Sanofi, TouchONCOLOGYResearch Funding: Merck (Inst), Merck KGaA (Inst)Other Relationship: GRiFOLS Josep TaberneroStock and Other Ownership Interests: Oniria TherapeuticsConsulting or Advisory Role: Bayer, Boehringer Ingelheim, Lilly, MSD, Merck Serono, Novartis, Sanofi, Taiho Pharmaceutical, Peptomyc, Chugai Pharma, Pfizer, Seattle Genetics, Array BioPharma, AstraZeneca, Genentech, Menarini, Servier, HalioDx, F. Hoffmann La Roche, Mirati Therapeutics, Pierre Fabre, Tessa Therapeutics, TheraMyc, Daiichi Sankyo, Samsung Bioepis, IQvia, Ikena Oncology, Merus, Neophore, Orion Biotechnology, Hutchison MediPharma, Scandion Oncology, Ona Therapeutics, Sotio, Inspirna, Scorpion TherapeuticsOther Relationship: Imedex, Medscape, MJH Life Sciences, Peerview, Physicans' Education Resource Alan SpatzConsulting or Advisory Role: Janssen, Bristol Myers Squibb, Pfizer/EMD Serono, Merck, Roche, AstraZeneca, Novartis, Bayer, AbbVieResearch Funding: Merck (Inst), Bristol Myers Squibb (Inst) C.S. PrameshStock and Other Ownership Interests: Aurobindo Philippe GirardHonoraria: Bayer, LEO Pharma, BMS/PfizerConsulting or Advisory Role: LEO Pharma, Bayer, BMS/PfizerTravel, Accommodations, Expenses: Bayer, LEO Pharma, BMS/Pfizer Jean-Yves BlayLeadership: Innate PharmaHonoraria: Roche, AstraZeneca, PharmaMar, MSD, BMS, Bayer, Ignyta, DecipheraConsulting or Advisory Role: Roche, Pharmamar, Deciphera, Blueprint Medicines, Bayer, Karyopharm TherapeuticsResearch Funding: GlaxoSmithKline (Inst), Pharmamar (Inst), Novartis (Inst), Bayer (Inst), Roche (Inst), BMS (Inst), MSD (Inst), Deciphera (Inst), AstraZeneca (Inst), OSE Pharma (Inst)Travel, Accommodations, Expenses: Roche Angel PorgadorStock and Other Ownership Interests: PooldiConsulting or Advisory Role: PiNKPatents, Royalties, Other Intellectual Property: Ben-Gurion University, Israel, several patents Eitan RubinStock and Other Ownership Interests: Pfizer Razelle KurzrockLeadership: CureMatch, CureMetrixStock and Other Ownership Interests: CureMatch, IDbyDNA, CureMetrixHonoraria: Roche, EUSA Pharma, NeoGenomics Laboratories, Biocom, NeoMed, Advanced Therapeutics, LEK, AACR, Chugai Pharma USA, Wiley, Merck, Pfizer, Meyer Consulting, Foundation Medicine, Turning Point Therapeutics, Bicara TherapeuticsConsulting or Advisory Role: Actuate Therapeutics, Loxo, XBiotech, NeoMed, Roche, Gaido, Soluventis, Pfizer, Merck, Turning Point Therapeutics, TD2/Volastra, Bicara Therapeutics, AstraZeneca, Biological Dynamics, Daiichi Sankyo, Eisai, EOM Pharmaceuticals, lylon, ProsperdtxSpeakers' Bureau: RocheResearch Funding: Guardant Health (Inst), Sequenom (Inst), Merck Serono (Inst), Genentech (Inst), Pfizer (Inst), Foundation Medicine (Inst), Incyte (Inst), Konica Minolta (Inst), Grifols (Inst), OmniSeq (Inst), Debiopharm Group (Inst), Boehringer Ingelheim (Inst), Top Alliance BioScience (Inst), Takeda (Isnt), Medlmmune (Inst), Biological Dynamics (Inst)Travel, Accommodations, Expenses: Roche, EUSA Pharma, NeoGenomics Laboratories, Biocom, NeoMed, Advanced Therapeutics, LEK, AACR, Chugai Pharma USA, Wiley Richard L. SchilskyLeadership: Clarified Precision MedicineStock and Other Ownership Interests: EQRxConsulting or Advisory Role: Cellworks, Scandion Oncology, Bryologyx, IIIumina, EQRx, Syapse, ZephyrAIResearch Funding: AstraZeneca (Inst), Bayer (Inst), Bristol Myers Squibb (Inst), Genentech/Roche (Inst), Lilly (Inst), Merck (Inst), Pfizer (Inst), Boehringer Ingelheim (Inst), Seattle Genetics (Inst)Open Payments Link: https://openpaymentsdata.cms.gov/physician/I138818/summaryNo other potential conflicts of interest were reported.
- Published
- 2022
- Full Text
- View/download PDF
23. A Non-Hazardous Deparaffinization Protocol Enables Quantitative Proteomics of Core Needle Biopsy-Sized Formalin-Fixed and Paraffin-Embedded (FFPE) Tissue Specimens.
- Author
-
Mitsa G, Guo Q, Goncalves C, Preston SEJ, Lacasse V, Aguilar-Mahecha A, Benlimame N, Basik M, Spatz A, Batist G, Miller WH Jr, Del Rincon SV, Zahedi RP, and Borchers CH
- Subjects
- Biopsy, Large-Core Needle, Formaldehyde, Humans, Paraffin Embedding, Proteome metabolism, Tissue Fixation, Paraffin, Proteomics methods
- Abstract
Most human tumor tissues that are obtained for pathology and diagnostic purposes are formalin-fixed and paraffin-embedded (FFPE). To perform quantitative proteomics of FFPE samples, paraffin has to be removed and formalin-induced crosslinks have to be reversed prior to proteolytic digestion. A central component of almost all deparaffinization protocols is xylene, a toxic and highly flammable solvent that has been reported to negatively affect protein extraction and quantitative proteome analysis. Here, we present a 'green' xylene-free protocol for accelerated sample preparation of FFPE tissues based on paraffin-removal with hot water. Combined with tissue homogenization using disposable micropestles and a modified protein aggregation capture (PAC) digestion protocol, our workflow enables streamlined and reproducible quantitative proteomic profiling of FFPE tissue. Label-free quantitation of FFPE cores from human ductal breast carcinoma in situ (DCIS) xenografts with a volume of only 0.79 mm
3 showed a high correlation between replicates (r2 = 0.992) with a median %CV of 16.9%. Importantly, this small volume is already compatible with tissue micro array (TMA) cores and core needle biopsies, while our results and its ease-of-use indicate that further downsizing is feasible. Finally, our FFPE workflow does not require costly equipment and can be established in every standard clinical laboratory.- Published
- 2022
- Full Text
- View/download PDF
24. Access Denied? The Unintended Consequences of Pending Drug Pricing Rules.
- Author
-
Kaplan A, Stewart DJ, Batist G, Spadafora S, Sehdev S, and Goodman SG
- Subjects
- Canada, Costs and Cost Analysis, Humans, Drug Costs
- Abstract
The government of Canada now plans to bring into force new federal drug pricing regulations on 1 July 2022. We do not take issue with the goal of medication affordability, which is vital in healthcare the world over. Our concern is that the new guidelines are being implemented without due consideration for three major unintended consequences: regulatory changes will lower the number of clinical trials for new medications in Canada, fewer clinical trials will mean lower research and development investments, and changes will reduce patients' access to new medications. Access to effective medications is a cornerstone of healthcare for Canadian patients. As physicians, our duty to patient care demands that we tell the government to protect the right of Canadians to timely access to life-changing medicines.
- Published
- 2022
- Full Text
- View/download PDF
25. Geriatric risk factors for serious COVID-19 outcomes among older adults with cancer: a cohort study from the COVID-19 and Cancer Consortium.
- Author
-
Elkrief A, Hennessy C, Kuderer NM, Rubinstein SM, Wulff-Burchfield E, Rosovsky RP, Vega-Luna K, Thompson MA, Panagiotou OA, Desai A, Rivera DR, Khaki AR, Tachiki L, Lynch RC, Stratton C, Elias R, Batist G, Kasi A, Shah DP, Bakouny Z, Cabal A, Clement J, Crowell J, Dixon B, Friese CR, Fry SL, Grover P, Gulati S, Gupta S, Hwang C, Khan H, Kim SJ, Klein EJ, Labaki C, McKay RR, Nizam A, Pennell NA, Puc M, Schmidt AL, Shahrokni A, Shaya JA, Su CT, Wall S, Williams N, Wise-Draper TM, Mishra S, Grivas P, French B, Warner JL, and Wildes TM
- Subjects
- Aged, COVID-19 Testing, Cohort Studies, Humans, Middle Aged, Risk Factors, SARS-CoV-2, COVID-19, Neoplasms
- Abstract
Background: Older age is associated with poorer outcomes of SARS-CoV-2 infection, although the heterogeneity of ageing results in some older adults being at greater risk than others. The objective of this study was to quantify the association of a novel geriatric risk index, comprising age, modified Charlson comorbidity index, and Eastern Cooperative Oncology Group performance status, with COVID-19 severity and 30-day mortality among older adults with cancer., Methods: In this cohort study, we enrolled patients aged 60 years and older with a current or previous cancer diagnosis (excluding those with non-invasive cancers and premalignant or non-malignant conditions) and a current or previous laboratory-confirmed COVID-19 diagnosis who reported to the COVID-19 and Cancer Consortium (CCC19) multinational, multicentre, registry between March 17, 2020, and June 6, 2021. Patients were also excluded for unknown age, missing data resulting in unknown geriatric risk measure, inadequate data quality, or incomplete follow-up resulting in unknown COVID-19 severity. The exposure of interest was the CCC19 geriatric risk index. The primary outcome was COVID-19 severity and the secondary outcome was 30-day all-cause mortality; both were assessed in the full dataset. Adjusted odds ratios (ORs) and 95% CIs were estimated from ordinal and binary logistic regression models., Findings: 5671 patients with cancer and COVID-19 were included in the analysis. Median follow-up time was 56 days (IQR 22-120), and median age was 72 years (IQR 66-79). The CCC19 geriatric risk index identified 2365 (41·7%) patients as standard risk, 2217 (39·1%) patients as intermediate risk, and 1089 (19·2%) as high risk. 36 (0·6%) patients were excluded due to non-calculable geriatric risk index. Compared with standard-risk patients, high-risk patients had significantly higher COVID-19 severity (adjusted OR 7·24; 95% CI 6·20-8·45). 920 (16·2%) of 5671 patients died within 30 days of a COVID-19 diagnosis, including 161 (6·8%) of 2365 standard-risk patients, 409 (18·5%) of 2217 intermediate-risk patients, and 350 (32·1%) of 1089 high-risk patients. High-risk patients had higher adjusted odds of 30-day mortality (adjusted OR 10·7; 95% CI 8·54-13·5) than standard-risk patients., Interpretation: The CCC19 geriatric risk index was strongly associated with COVID-19 severity and 30-day mortality. Our CCC19 geriatric risk index, based on readily available clinical factors, might provide clinicians with an easy-to-use risk stratification method to identify older adults most at risk for severe COVID-19 as well as mortality., Funding: US National Institutes of Health National Cancer Institute Cancer Center., Competing Interests: The following authors declare competing interests not related to the current work: ZB reports grants from Genentech/imCORE, non-financial support from Bristol Myers Squibb, and personal fees from UpToDate. AE reports salary support from the Canadian Institute of Health Research, the Detweiler Travelling Fellowship (Royal College of Physicians and Surgeons of Canada), and the Henry R Shibata Fellowship (Cedar's Cancer Foundation). CRF reports grants from Merck Foundation, NCCN/Pfizer, and National Cancer Institute, and other support from National Cancer Institute and the Patient-Centered Outcomes Research Institute. PGri reports personal fees and non-financial support from AstraZeneca, personal fees from Astellas Pharma, personal fees from Bayer, grants and personal fees from Bristol Myers Squibb, grants and non-financial support from Clovis Oncology, personal fees from Dyania Health, grants and personal fees from EMD Serono, personal fees from Exelixis, personal fees from Foundation Medicine, personal fees from Genentech/Roche, personal fees from Genzyme, grants and personal fees from GlaxoSmithKline, personal fees from Guardant Health, grants and personal fees from Immunomedics/Gilead, personal fees from Infinity Pharmaceuticals, personal fees from Janssen, grants and personal fees from Merck, grants and personal fees from Mirati Therapeutics, grants and personal fees from Pfizer, grants and personal fees from QED Therapeutics, personal fees from Regeneron Pharmaceuticals, personal fees from Seattle Genetics, personal fees from 4D Pharma, personal fees from UroGen, grants from Bavarian Nordic, and grants from Debiopharm. SGup reports grants and personal fees from Bristol Myers Squibb, personal fees from Merck, Janssen, Seattle Genetis, EMD Sorono, and Pfizer, and grants from Astellas and BMS. CHw reports grants from Merck, Bayer, and AstraZeneca, and personal fees from Tempus and EMD Sorono, and other support from Johnson and Johnson. AK reports other support from TESARO, Fibrogen, Geistlich Pharma, Astellas Pharma, Rafael Pharmaceuticals, and Novocure. ARK reports other support from Merck and Sanofi, personal fees from OncLive, grants from ASCO Conquer Cancer Foundation, and grants from Bladder Cancer Advocacy Network. HK reports position on advisory board of Sanofi Genzyme NSCLC Northeast. NMK reports personal fees from BMS, Janssen, Seattle Genetics, Celldex, Sandoz, Invitae, Beyond Spring, Spectrum G1 Therapeutics, and Total Health, and grants from Amgen, Jazz Therapeutics, G1 Therapeutics, and Samsung. CL reports grants from imCORE/Genentech. RRM reports serving on advisory board or as a consultant for Astrazeneca, Aveo, Bayer, BMS, Caris, Dendreon, Exelixis, Janssen, Merck, Myovant, Novartis, Pfizer, Sanofi, Sorrento Therapeutics, and Tempus. RRM received institutional research funding from Pfizer, Bayer, Tempus. SM reports personal fees from National Geographic. OAP reports grants from National Institutes of Health (NIH) and Agency for Healthcare Research and Quality, and personal fees from International Consulting Associates. NAP reports personal fees from AstraZeneca, Merck, Pfizer, Eli Lilly, Genentech, BMS, Amgen, Inivata, G1 Therapeutics, Xencor, Mirati, Janssen, Boehringer Ingelheim, and Sanofi-Genzyme-Regeneron. RPR reports grants from BMS and Janssen, and personal fees from BMS, Janssen, Dova, and Inari. MAT reports personal fees from VIA Oncology, GSK, and Adaptive Advisory Board, other support from Syapse, UpToDate, Takeda, Celgene, Doximity, AbbVie, BMS, CRAB CTC, Denovo, Hoosier Research Network, Lilly, LynxBio, Strata Oncology, and TG Therapeutics. JLW reports personal fees from Roche, Westat, Flatiron Health, Melax Tech, and IBM Watson Health, other support from HemOnc and Janssen, and grants from AACR. TMW reports personal fees from Carevive, and personal fees from Sanofi, and Seattle Genetics. TMW-D reports grants from BMS, Merck, Janssen, and GSK/Tesaro, personal fees from Exicure, Shattuck Labs, Merck, Caris Life Science, and SITC, and other support from High Enroll. EW-B reports grants from Pfizer Global Medical Grants, personal fees from Astellas, Aveo Oncology Bristol Myers Squibb, Exelixis, and Janssen, and other support from Immunomedics and Nektar. The following authors declare competing interests during the conduct of the study: SM reports grants and other support from National Cancer Institute and from the International Association for the Study of Lung Cancer. DPS reports grants from American Cancer Society and Hope Foundation for Cancer Research and from NIH. LT reports grants from NIH. JLW reports grants from NIH. All other authors declare no competing interests., (© 2022 The Author(s). Published by Elsevier Ltd. This is an Open Access article under the CC BY-NC-ND 4.0 license.)
- Published
- 2022
- Full Text
- View/download PDF
26. Treatment Access, Health Economics, and the Wave of a Magic Wand.
- Author
-
Stewart DJ, Bradford JP, and Batist G
- Subjects
- Canada, Costs and Cost Analysis, Humans, Quality-Adjusted Life Years, Drug Costs
- Abstract
New drugs are expensive, in part due to excessive drug development costs. Governments are trying to reduce drug prices. This can delay access to effective agents. A country's access to new drugs correlates with prices they agree to pay. After Health Canada approves a drug, the Canadian Agency for Drug and Technologies in Health (CADTH) assesses it. CADTH's approval is usually contingent on it costing ≤CAD 50,000 per quality adjusted life year (QALY) gained. This value (unchanged from the 1970s) is inappropriately low. An inflation-adjusted CAD 50,000 1975 QALY should translate into a CAD 250,000 2021 QALY. CADTH's target also does not consider that drug development costs have risen much faster than inflation or that new precision therapies may only be used in small populations. In a separate process, proposals from the Patented Medicines Price Review Board (PMPRB) would decrease initial Canadian drug prices by 20%, but prices would fall further as sales increased, with ultimate price reductions of up to 80%. PMPRB claims its proposal would not reduce drug access, but multiple analyses strongly suggest otherwise. Government price controls target the symptom (high prices), not the disease. They translate into shortages without solving the problem. CADTH and PMPRB approaches both threaten access to effective drugs.
- Published
- 2022
- Full Text
- View/download PDF
27. Immuno-MALDI-MS for Accurate Quantitation of Targeted Peptides from Volume-Restricted Samples.
- Author
-
Sobsey CA, Froehlich B, Batist G, and Borchers CH
- Subjects
- Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization methods, Peptides analysis, Proteins analysis
- Abstract
The immuno-MALDI-MS method can be used to quantify low-abundance proteins from clinical samples that offer only a limited amount of material for analysis. An internal standard, in the form of a stable isotope-labeled peptide, is used to ensure reproducible and absolute quantitation. The protocol described here was optimized for the quantitation of AKT1 and AKT2, but we offer instructions on how to adapt the method to target other proteins. The described workflow is compatible with automation via a liquid handling robot for high-throughput applications., (© 2022. The Author(s), under exclusive license to Springer Science+Business Media, LLC, part of Springer Nature.)
- Published
- 2022
- Full Text
- View/download PDF
28. A multiplexed, automated immuno-matrix assisted laser desorption/ionization mass spectrometry assay for simultaneous and precise quantitation of PTEN and p110α in cell lines and tumor tissues.
- Author
-
Froehlich BC, Popp R, Sobsey CA, Ibrahim S, LeBlanc A, Mohammed Y, Buchanan M, Aguilar-Mahecha A, Pötz O, Chen MX, Spatz A, Basik M, Batist G, Zahedi RP, and Borchers CH
- Subjects
- Cell Line, Tumor, Female, Humans, Lasers, Neoplasm Proteins, PTEN Phosphohydrolase, Reproducibility of Results, Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization, Biomarkers, Tumor, Breast Neoplasms
- Abstract
The PI3-kinase/AKT/mTOR pathway plays a central role in cancer signaling. While p110α is the catalytic α-subunit of PI3-kinase and a major drug target, PTEN is the main negative regulator of the PI3-kinase/AKT/mTOR pathway. PTEN is often down-regulated in cancer, and there are conflicting data on PTEN's role as breast cancer biomarker. PTEN and p110α protein expression in tumors is commonly analyzed by immunohistochemistry, which suffers from poor multiplexing capacity, poor standardization, and antibody crossreactivity, and which provides only semi-quantitative data. Here, we present an automated, and standardized immuno-matrix-assisted laser desorption/ionization mass spectrometry (iMALDI) assay that allows precise and multiplexed quantitation of PTEN and p110α concentrations, without the limitations of immunohistochemistry. Our iMALDI assay only requires a low-cost benchtop MALDI-TOF mass spectrometer, which simplifies clinical translation. We validated our assay's precision and accuracy, with simultaneous enrichment of both target proteins not significantly affecting the precision and accuracy of the quantitation when compared to the PTEN- and p110α-singleplex iMALDI assays (<15% difference). The multiplexed assay's linear range is from 0.6-20 fmol with accuracies of 90-112% for both target proteins, and the assay is free of matrix-related interferences. The inter-day reproducibility over 5-days was high, with an overall CV of 9%. PTEN and p110α protein concentrations can be quantified down to 1.4 fmol and 0.6 fmol per 10 μg of total tumor protein, respectively, in various tumor tissue samples, including fresh-frozen breast tumors and colorectal cancer liver metastases, and patient-derived xenograft (PDX) tumors.
- Published
- 2021
- Full Text
- View/download PDF
29. CRAFT-A Proposed Framework for Decentralized Clinical Trials Participation in Canada.
- Author
-
Sundquist S, Batist G, Brodeur-Robb K, Dyck K, Eigl BJ, Lee DK, Limoges J, Longstaff H, Pankovich J, Sadura A, Sullivan P, and Dancey JE
- Subjects
- Canada, Delivery of Health Care, Humans, Rural Population, Telemedicine
- Abstract
Canada's vast geography, and centralized delivery of cancer care and clinical trials create barriers for trial participation for patients in remote and rural settings. The development and implementation of a framework that enables safe and regulatory compliant trial participation through local healthcare providers would benefit Canadian patients, clinicians, trial sponsors and the health care system. To address this issue, representatives of Canada's cancer clinical trial community met to identify key challenges and develop recommendations for remote patient participation in trials. A structured literature review identified remote/rural trial delivery models. A panel of expert stakeholders reviewed the models and participated in a workshop to assess health system readiness, identify needed processes, tools and mechanisms, and develop recommendations for a Canadian framework for decentralized clinical trial conduct. The Canadian Remote Access Framework for clinical Trials (CRAFT) represents a risk-based approach used by site investigators to delegate responsibilities for a given trial to satellite health centres within a hub-and-spoke "trial cluster". The Framework includes specific recommendations to ensure research experience, capacity, regulatory compliance and patient safety. Canada's cancer care and telemedicine systems can be leveraged to enable broader access to clinical trials for patients who are geographically remote from cancer centres. CRAFT's risk-based framework is based on other successful models of remote trial patient management and is in the pilot implementation phase in Canada.
- Published
- 2021
- Full Text
- View/download PDF
30. Precise Quantitation of PTEN by Immuno-MRM: A Tool To Resolve the Breast Cancer Biomarker Controversy.
- Author
-
Ibrahim S, Lan C, Chabot C, Mitsa G, Buchanan M, Aguilar-Mahecha A, Elchebly M, Poetz O, Spatz A, Basik M, Batist G, Zahedi RP, and Borchers CH
- Subjects
- Female, Humans, Immunohistochemistry, PTEN Phosphohydrolase, Phosphatidylinositol 3-Kinases, Biomarkers, Tumor, Breast Neoplasms diagnosis
- Abstract
The tumor suppressor PTEN is the main negative regulator of PI3K/AKT/mTOR signaling and is commonly found downregulated in breast cancer (BC). Conflicting data from conventional immunoassays such as immunohistochemistry (IHC) has sparked controversy about PTEN's role as a prognostic and predictive biomarker in BC, which can be largely attributed to the lack of specificity, sensitivity, and interlaboratory standardization. Here, we present a fully standardized, highly sensitive, robust microflow immuno-MRM (iMRM) assay that enables precise quantitation of PTEN concentrations in cells and fresh frozen (FF) and formalin-fixed paraffin-embedded (FFPE) tissues, down to 0.1 fmol/10 μg of extracted protein, with high interday and intraday precision (CV 6.3%). PTEN protein levels in BC PDX samples that were determined by iMRM correlate well with semiquantitative IHC and WB data. iMRM, however, allowed the precise quantitation of PTEN-even in samples that were deemed to be PTEN negative by IHC or western blot (WB)-while requiring substantially less tumor tissue than WB. This is particularly relevant because the extent of PTEN downregulation in tumors has been shown to correlate with severity. Our standardized and robust workflow includes an 11 min microflow LC-MRM analysis on a triple-quadrupole MS and thus provides a much needed tool for the study of PTEN as a potential biomarker for BC.
- Published
- 2021
- Full Text
- View/download PDF
31. Aurora kinase A inhibitor, LY3295668 erbumine: a phase 1 monotherapy safety study in patients with locally advanced or metastatic solid tumors.
- Author
-
Chu QS, Bouganim N, Fortier C, Zaknoen S, Stille JR, Kremer JD, Yuen E, Hui YH, de la Peña A, Lithio A, Smith PS, and Batist G
- Subjects
- Adult, Aged, Dose-Response Relationship, Drug, Female, Humans, Male, Maximum Tolerated Dose, Middle Aged, Neoplasms pathology, Piperidines adverse effects, Protein Kinase Inhibitors adverse effects, Pyrazoles adverse effects, Treatment Outcome, Aurora Kinase A antagonists & inhibitors, Neoplasms drug therapy, Piperidines therapeutic use, Protein Kinase Inhibitors therapeutic use, Pyrazoles therapeutic use
- Abstract
Background Aurora A kinase (AurA) overexpression likely contributes to tumorigenesis and therefore represents an attractive target for cancer therapeutics. This phase 1 study aimed to determine the safety, pharmacokinetics, and antitumor activity of LY3295668 erbumine, an AurA inhibitor, in patients with locally advanced or metastatic solid tumors. Methods Patients with locally advanced or metastatic solid tumors, Eastern Cooperative Oncology Group performance status 0-1, and disease progression after one to four prior treatment regimens were enrolled. Primary objective was to determine maximum tolerated dose (MTD); secondary objectives included evaluation of the tolerability and safety profile and pharmacokinetics of LY3295668. All patients received twice-daily (BID) oral LY3295668 in 21-day cycles in an ascending-dose schedule. Results Twelve patients were enrolled in phase 1 (25 mg, n = 8; 50 mg, n = 2; 75 mg, n = 2) and one patient was enrolled after. Overall, four patients experienced dose-limiting toxicities (DLTs) within the first cycle (75 mg: Grade 3 diarrhea [one patient], Grade 4 mucositis and Grade 3 corneal deposits [one patient]; 50 mg: mucositis and diarrhea [both Grade 3, one patient]; 25 mg: Grade 3 mucositis [one patient]). Patients exhibiting DLTs had the highest model-predicted exposures at steady state. Mucositis was the most common adverse event (67%), followed by diarrhea, fatigue, alopecia, anorexia, constipation, and nausea. Nine patients had best response of stable disease; the disease control rate was 69%. Conclusions MTD of LY3295668 was 25 mg BID. LY3295668 had a manageable toxicity profile and demonstrated activity in some patients with locally advanced or metastatic solid tumors.Trial registration ClinicalTrials.gov, NCT03092934. Registered March 22, 2017. https://clinicaltrials.gov/ct2/show/NCT03092934 ., (© 2021. The Author(s), under exclusive licence to Springer Science+Business Media, LLC part of Springer Nature.)
- Published
- 2021
- Full Text
- View/download PDF
32. Copresence of High-Risk Human Papillomaviruses and Epstein-Barr Virus in Colorectal Cancer: A Tissue Microarray and Molecular Study from Lebanon.
- Author
-
Nagi K, Gupta I, Jurdi N, Yasmeen A, Vranic S, Batist G, and Moustafa AA
- Subjects
- Adolescent, Adult, Aged, Aged, 80 and over, Case-Control Studies, Child, Child, Preschool, Cohort Studies, Colorectal Neoplasms virology, DNA, Viral genetics, Epstein-Barr Virus Infections virology, Female, Humans, Immunohistochemistry, Lebanon epidemiology, Male, Middle Aged, Papillomavirus Infections virology, Polymerase Chain Reaction, Prevalence, Risk Factors, Young Adult, Alphapapillomavirus genetics, Colorectal Neoplasms epidemiology, Epstein-Barr Virus Infections epidemiology, Herpesvirus 4, Human genetics, Papillomavirus Infections epidemiology
- Abstract
Colorectal cancer (CRC) is the third most common cause of cancer-related deaths worldwide. Human papillomaviruses (HPVs) and Epstein-Barr virus (EBV) have been reported to be present in different types of human cancers, including CRCs, where they can play a key role in the onset and/or progression of these cancers. Thus, we herein explored the prevalence of high-risk HPVs and EBV in a cohort of 94 CRC tissue samples and 13 colorectal normal tissues from the Lebanese population using polymerase chain reaction, immunohistochemistry, and tissue microarray methodologies. We found that high-risk HPVs are present in 64%, while EBV is present in 29% of our CRC samples. Additionally, our data showed that high-risk HPV types (16, 18, 35, 58, 51, 45, 52, 31, and 33) are the most frequent in CRC in the Lebanese cohort, respectively. Our data point out that HPVs and EBV are copresent in 28% of the samples. Thus, this study clearly suggests that high-risk HPVs and EBV are present/copresent in CRCs, where they could play an important role in colorectal carcinogenesis. Nevertheless, further investigations using a larger cohort are needed to elucidate the possible cooperation between these oncoviruses in the development of CRC.
- Published
- 2021
- Full Text
- View/download PDF
33. Site-Specific Variation in Radiomic Features of Head and Neck Squamous Cell Carcinoma and Its Impact on Machine Learning Models.
- Author
-
Liu X, Maleki F, Muthukrishnan N, Ovens K, Huang SH, Pérez-Lara A, Romero-Sanchez G, Bhatnagar SR, Chatterjee A, Pusztaszeri MP, Spatz A, Batist G, Payabvash S, Haider SP, Mahajan A, Reinhold C, Forghani B, O'Sullivan B, Yu E, and Forghani R
- Abstract
Current radiomic studies of head and neck squamous cell carcinomas (HNSCC) are typically based on datasets combining tumors from different locations, assuming that the radiomic features are similar based on histopathologic characteristics. However, molecular pathogenesis and treatment in HNSCC substantially vary across different tumor sites. It is not known if a statistical difference exists between radiomic features from different tumor sites and how they affect machine learning model performance in endpoint prediction. To answer these questions, we extracted radiomic features from contrast-enhanced neck computed tomography scans (CTs) of 605 patients with HNSCC originating from the oral cavity, oropharynx, and hypopharynx/larynx. The difference in radiomic features of tumors from these sites was assessed using statistical analyses and Random Forest classifiers on the radiomic features with 10-fold cross-validation to predict tumor sites, nodal metastasis, and HPV status. We found statistically significant differences ( p -value ≤ 0.05) between the radiomic features of HNSCC depending on tumor location. We also observed that differences in quantitative features among HNSCC from different locations impact the performance of machine learning models. This suggests that radiomic features may reveal biologic heterogeneity complementary to current gold standard histopathologic evaluation. We recommend considering tumor site in radiomic studies of HNSCC.
- Published
- 2021
- Full Text
- View/download PDF
34. High-risk human papillomaviruses and Epstein-Barr virus in breast cancer in Lebanese women and their association with tumor grade: a molecular and tissue microarray study.
- Author
-
Nagi K, Gupta I, Jurdi N, Jabeen A, Yasmeen A, Batist G, Vranic S, and Al-Moustafa AE
- Abstract
Background: High-risk human papillomaviruses (HPVs) are present and can cooperate with Epstein-Barr virus (EBV) to initiate and/or enhance the progression of several types of human carcinomas including cervical as well as head and neck; in parallel, it has been recently pointed out that these oncoviruses can be detected in human breast cancers. Thus, we herein explored the presence/co-presence of high-risk HPVs and EBV in breast cancer in Lebanese women., Methods: A cohort of 102 breast cancer samples and 14 normal breast tissues were assessed for the presence of HPVs and EBV. Polymerase chain reaction (PCR) and immunohistochemistry (IHC) analysis in addition to tissue microarray (TMA) platform were used in this study., Results: We found the presence of HPV in 66/102 (65%) of our samples, while EBV is present in 41/102 (40%) of the cohort. Additionally, our data showed that high-risk HPV types (52, 35, 58, 45, 16 and 51) are the most frequent in breast cancer in Lebanese women. Meanwhile, we report that high-risk HPVs and EBV are co-present in 30/102 (29%) of the samples; more significantly, our results indicate that their co-presence is associated with tumor grade (p = 0.03)., Conclusion: Our data revealed that HPVs and EBV are present/co-present in human breast cancer where they may play an important role in its development and/or progression; thus, we believe that further investigations are essential to confirm and elucidate the presence/co-presence of these oncoviruses and the underlying mechanisms of their interaction in breast carcinogenesis.
- Published
- 2021
- Full Text
- View/download PDF
35. Why Tumor Genetic Heterogeneity May Require Rethinking Cancer Genesis and Treatment.
- Author
-
Gottlieb B, Trifiro M, and Batist G
- Subjects
- Aging genetics, Antineoplastic Agents therapeutic use, Carcinogenesis drug effects, Carcinogenesis pathology, Drug Resistance, Neoplasm genetics, Humans, Neoplasms genetics, Neoplasms pathology, Phenotype, Tumor Microenvironment drug effects, Tumor Microenvironment genetics, Antineoplastic Agents pharmacology, Carcinogenesis genetics, Genetic Heterogeneity, Neoplasms drug therapy
- Abstract
Tumor genetic heterogeneity, in which individual tumors contain both multiple variant cancer-associated and normal genes, has been widely reported, although its significance has yet to be fully understood. We propose a genetic heterogeneity-based selection-centric hypothesis in which genetic heterogeneity, caused by the temporary reduction of DNA repair efficiency, occurs very early in human development, resulting in a small minority of cells in normal tissues acquiring cancer-associated genes that remain dormant. Cancer develops when precancer cells are selected for by altered tissue microenvironments; similar scenarios occur with development of metastases and therapeutic resistance in established cancer. This suggests that a normal cell selection treatment approach based on preferentially selecting normal cells within tumors may be effective in treating cancer., (Copyright © 2020 Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
36. Digital Display Precision Predictor: the prototype of a global biomarker model to guide treatments with targeted therapy and predict progression-free survival.
- Author
-
Lazar V, Magidi S, Girard N, Savignoni A, Martini JF, Massimini G, Bresson C, Berger R, Onn A, Raynaud J, Wunder F, Berindan-Neagoe I, Sekacheva M, Braña I, Tabernero J, Felip E, Porgador A, Kleinman C, Batist G, Solomon B, Tsimberidou AM, Soria JC, Rubin E, Kurzrock R, and Schilsky RL
- Abstract
The expanding targeted therapy landscape requires combinatorial biomarkers for patient stratification and treatment selection. This requires simultaneous exploration of multiple genes of relevant networks to account for the complexity of mechanisms that govern drug sensitivity and predict clinical outcomes. We present the algorithm, Digital Display Precision Predictor (DDPP), aiming to identify transcriptomic predictors of treatment outcome. For example, 17 and 13 key genes were derived from the literature by their association with MTOR and angiogenesis pathways, respectively, and their expression in tumor versus normal tissues was associated with the progression-free survival (PFS) of patients treated with everolimus or axitinib (respectively) using DDPP. A specific eight-gene set best correlated with PFS in six patients treated with everolimus: AKT2, TSC1, FKB-12, TSC2, RPTOR, RHEB, PIK3CA, and PIK3CB (r = 0.99, p = 5.67E-05). A two-gene set best correlated with PFS in five patients treated with axitinib: KIT and KITLG (r = 0.99, p = 4.68E-04). Leave-one-out experiments demonstrated significant concordance between observed and DDPP-predicted PFS (r = 0.9, p = 0.015) for patients treated with everolimus. Notwithstanding the small cohort and pending further prospective validation, the prototype of DDPP offers the potential to transform patients' treatment selection with a tumor- and treatment-agnostic predictor of outcomes (duration of PFS).
- Published
- 2021
- Full Text
- View/download PDF
37. Selective AKT kinase inhibitor capivasertib in combination with fulvestrant in PTEN-mutant ER-positive metastatic breast cancer.
- Author
-
Smyth LM, Batist G, Meric-Bernstam F, Kabos P, Spanggaard I, Lluch A, Jhaveri K, Varga A, Wong A, Schram AM, Ambrose H, Carr TH, de Bruin EC, Salinas-Souza C, Foxley A, Hauser J, Lindemann JPO, Maudsley R, McEwen R, Moschetta M, Nikolaou M, Schiavon G, Razavi P, Banerji U, Baselga J, Hyman DM, and Chandarlapaty S
- Abstract
Five to ten percent of ER+ metastatic breast cancer (MBC) tumors harbor somatic PTEN mutations. Loss of function of this tumor-suppressor gene defines a highly aggressive, treatment-refractory disease for which new therapies are urgently needed. This Phase I multipart expansion study assessed oral capivasertib with fulvestrant in patients with PTEN-mutant ER+ MBC. Safety and tolerability were assessed by standard methods. Plasma and tumor were collected for NGS and immunohistochemistry analyses of PTEN protein expression. In 31 eligible patients (12 fulvestrant naive; 19 fulvestrant pretreated), the 24-week clinical benefit rate was 17% in fulvestrant-naive and 42% in fulvestrant-pretreated patients, with objective response rate of 8% and 21%, respectively. Non-functional PTEN was centrally confirmed in all cases by NGS or immunohistochemistry. Co-mutations occurred in PIK3CA (32%), with less ESR1 (10% vs 72%) and more TP53 (40% vs 28%) alterations in fulvestrant-naive versus fulvestrant-pretreated patients, respectively. PTEN was clonally dominant in most patients. Treatment-related grade ≥3 adverse events occurred in 32% of patients, most frequently diarrhea and maculopapular rash (both n = 2). In this clinical study, which selectively targeted the aggressive PTEN-mutant ER+ MBC, capivasertib plus fulvestrant was tolerable and clinically active. Phenotypic and genomic differences were apparent between fulvestrant-naive and -pretreated patients.Trial registration number for the study is NCT01226316.
- Published
- 2021
- Full Text
- View/download PDF
38. Copy number and transcriptome alterations associated with metastatic lesion response to treatment in colorectal cancer.
- Author
-
Gambaro K, Marques M, McNamara S, Couetoux du Tertre M, Diaz Z, Hoffert C, Srivastava A, Hébert S, Samson B, Lespérance B, Ko YJ, Dalfen R, St-Hilaire E, Sideris L, Couture F, Burkes R, Harb M, Camlioglu E, Gologan A, Pelsser V, Constantin A, Greenwood CMT, Tejpar S, Kavan P, Kleinman CL, and Batist G
- Subjects
- Adult, Aged, Aged, 80 and over, Antineoplastic Agents therapeutic use, Bevacizumab therapeutic use, Colorectal Neoplasms drug therapy, Colorectal Neoplasms pathology, Drug Resistance, Neoplasm genetics, Female, Gene Expression Regulation, Neoplastic, Humans, Liver Neoplasms secondary, Male, Middle Aged, Progression-Free Survival, Exome Sequencing, Colorectal Neoplasms genetics, DNA Copy Number Variations genetics, Transcriptome genetics
- Abstract
Background: Therapeutic resistance is the main cause of death in metastatic colorectal cancer. To investigate genomic plasticity, most specifically of metastatic lesions, associated with response to first-line systemic therapy, we collected longitudinal liver metastatic samples and characterized the copy number aberration (CNA) landscape and its effect on the transcriptome., Methods: Liver metastatic biopsies were collected prior to treatment (pre, n = 97) and when clinical imaging demonstrated therapeutic resistance (post, n = 43). CNAs were inferred from whole exome sequencing and were correlated with both the status of the lesion and overall patient progression-free survival (PFS). We used RNA sequencing data from the same sample set to validate aberrations as well as independent datasets to prioritize candidate genes., Results: We identified a significantly increased frequency gain of a unique CN, in liver metastatic lesions after first-line treatment, on chr18p11.32 harboring 10 genes, including TYMS, which has not been reported in primary tumors (GISTIC method and test of equal proportions, FDR-adjusted p = 0.0023). CNA lesion profiles exhibiting different treatment responses were compared and we detected focal genomic divergences in post-treatment resistant lesions but not in responder lesions (two-tailed Fisher's Exact test, unadjusted p ≤ 0.005). The importance of examining metastatic lesions is highlighted by the fact that 15 out of 18 independently validated CNA regions found to be associated with PFS in this study were only identified in the metastatic lesions and not in the primary tumors., Conclusion: This investigation of genomic-phenotype associations in a large colorectal cancer liver metastases cohort identified novel molecular features associated with treatment response, supporting the clinical importance of collecting metastatic samples in a defined clinical setting., (© 2021 The Authors. Clinical and Translational Medicine published by John Wiley & Sons Australia, Ltd on behalf of Shanghai Institute of Clinical Bioinformatics.)
- Published
- 2021
- Full Text
- View/download PDF
39. Optimizing treatment sequencing of chemotherapy for patients with rectal cancer: The KIR randomized phase II trial.
- Author
-
Garant A, Kavan P, Martin AG, Azoulay L, Vendrely V, Lavoie C, Vasilevsky CA, Boutros M, Faria J, Nguyen TN, Ferland E, Des Groseilliers S, Cloutier AS, Diec H, Drolet S, Richard C, Batist G, and Vuong T
- Subjects
- Chemotherapy, Adjuvant, Disease-Free Survival, Fluorouracil therapeutic use, Humans, Middle Aged, Neoadjuvant Therapy, Neoplasm Recurrence, Local pathology, Neoplasm Staging, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Rectal Neoplasms pathology
- Abstract
Background: Randomized studies have shown low compliance to adjuvant chemotherapy in rectal cancer patients receiving preoperative chemotherapy and external beam radiation (CT/EBRT) with total mesorectal excision. We hypothesize that giving neoadjuvant CT before local treatment would improve CT compliance., Methods: Between 2010-2017, 180 patients were randomized (2:1) to either Arm A (AA) with FOLFOX x6 cycles prior to high dose rate brachytherapy (HDRBT) and surgery plus adjuvant FOLFOX x6 cycles, or Arm B (AB), with neoadjuvant HDRBT with surgery and adjuvant FOLFOX x12 cycles. The primary endpoint was CT compliance to ≥85% of full-dose CT for the first six cycles. Secondary endpoints were ypT0N0, five-year disease free survival (DFS), local control and overall survival (OS)., Results: Patients were randomized to either AA (n = 120, median age (MA) 62 years) or AB (n = 60, MA 63 years). 175/180 patients completed HDRBT as planned (97.2%). In AA, two patients expired during CT; three patients post-randomization received short course EBRT because of progression under CT (n = 2, AA) or personal preference (n = 1, AB). ypT0N0 was 31% in AA and 28% in AB (p = 0.7). CT Compliance was 80% in AA and 53% in AB (p = 0.0002). Acute G3/G4 toxicity was 35.8% in AA and 27.6% in AB (p = 0.23). With a median follow-up of 48.5 months (IQR 33-72), the five-year DFS was 72.3% with AA and 68.3% with AB (p = 0.74), the five-year OS 83.8% for AA and 82.2% for AB (p = 0.53), and the five-year local recurrence was 6.3% for AA and 5.8% for AB (p = 0.71)., Conclusion: We confirmed improved compliance to neoadjuvant CT in this study. Although there is no statistical difference in ypT0N0 rate, local recurrence, and DFS between the two arms, a trend towards favourable oncological outcomes is observed., (Copyright © 2020 Elsevier B.V. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
40. Lung Cancer Pre-Diagnostic Pathways from First Presentation to Specialist Referral.
- Author
-
Khare SR, Madathil SA, Batist G, Peter Brojde Lung Cancer Group, and Vedel I
- Subjects
- Emergency Service, Hospital, Humans, Primary Health Care, Referral and Consultation, Retrospective Studies, Lung Neoplasms diagnosis
- Abstract
Background: Lung cancer is often diagnosed at a late stage with high associated mortality. Timely diagnosis depends on timely referral to a respiratory specialist; however, in Canada, little is known about how patients move through primary care to get to a respiratory specialist. Accordingly, we aimed to identify and describe lung cancer pre-diagnostic pathways in primary care from first presentation to referral., Methods: In this retrospective cohort study, patients with primary lung cancer were recruited using consecutive sampling ( n = 50) from a lung cancer center in Montréal, Québec. Data on healthcare service utilization in primary care were collected from chart reviews and structured patient interviews and analyzed using latent class analysis to identify groups of patients with similar pre-diagnostic pathways. Each group was described based on patient- and tumor-related characteristics and the sequence of utilization activities., Results: 68% of the patients followed a pathway where family physician (FP) visits were dominant ("FP-centric") and 32% followed a pathway where walk-in clinic and emergency department (ED) visits were dominant ("ED-centric"). Time to referral in the FP group was double that of the ED group (45 days (IQR: 12-111) vs. 22 (IQR: 5-69)) with more advanced disease (65% vs. 50%). In the FP group, 29% of the patients saw their FP three times or more before being referred and 41% had an ED visit., Conclusions: Our findings may reflect the challenge of diagnosing lung cancer in primary care, missed opportunities for earlier diagnosis, and a lack of integration between primary and specialist care.
- Published
- 2021
- Full Text
- View/download PDF
41. High mortality among hospital-acquired COVID-19 infection in patients with cancer: A multicentre observational cohort study.
- Author
-
Elkrief A, Desilets A, Papneja N, Cvetkovic L, Groleau C, Lakehal YA, Shbat L, Richard C, Malo J, Belkaid W, Cook E, Doucet S, Tran TH, Jao K, Daaboul N, Bhang E, Loree JM, Miller WH Jr, Vinh DC, Bouganim N, Batist G, Letendre C, and Routy B
- Subjects
- Adolescent, Adult, Aged, Aged, 80 and over, COVID-19, Canada epidemiology, Child, Child, Preschool, Cohort Studies, Coronavirus Infections complications, Coronavirus Infections virology, Female, Humans, Male, Middle Aged, Neoplasms epidemiology, Neoplasms virology, Pandemics, Pneumonia, Viral complications, Pneumonia, Viral virology, Prognosis, Risk Factors, SARS-CoV-2, Survival Rate, Young Adult, Betacoronavirus isolation & purification, Coronavirus Infections mortality, Coronavirus Infections transmission, Hospitals statistics & numerical data, Mortality trends, Neoplasms mortality, Pneumonia, Viral mortality, Pneumonia, Viral transmission
- Abstract
Introduction: Studies suggest that patients with cancer are more likely to experience severe outcomes from COVID-19. Therefore, cancer centres have undertaken efforts to care for patients with cancer in COVID-free units. Nevertheless, the frequency and relevance of nosocomial transmission of COVID-19 in patients with cancer remain unknown. The goal of this study was to determine the incidence and impact of hospital-acquired COVID-19 in this population and identify predictive factors for COVID-19 severity in patients with cancer., Methods: Patients with cancer and a laboratory-confirmed diagnosis of COVID-19 were prospectively identified using provincial registries and hospital databases between March 3rd and May 23rd, 2020 in the provinces of Quebec and British Columbia in Canada. Patient's baseline characteristics including age, sex, comorbidities, cancer type and type of anticancer treatment were collected. The exposure of interest was incidence of hospital-acquired infection defined by diagnosis of SARS-CoV-2 ≥ 5 days after hospital admission for COVID-unrelated cause. Co-primary outcomes were death or composite outcomes of severe illness from COVID-19 such as hospitalisation, supplemental oxygen, intensive-care unit (ICU) admission and/or mechanical ventilation., Results: A total of 252 patients (N = 249 adult and N = 3 paediatric) with COVID-19 and cancer were identified, and the majority were residents of Quebec (N = 233). One hundred and six patients (42.1%) received active anticancer treatment in the last 3 months before COVID-19 diagnosis. During a median follow-up of 25 days, 33 (13.1%) required admission to the ICU, and 71 (28.2%) died. Forty-seven (19.1%) had a diagnosis of hospital-acquired COVID-19. Median overall survival was shorter in those with hospital-acquired infection than that in a contemporary community-acquired population (27 days versus unreached, hazard ratio (HR) = 2.3, 95% CI: 1.2-4.4, p = 0.0006. Multivariate analysis demonstrated that hospital-acquired COVID-19, age, Eastern Cooperative Oncology Group status and advanced stage of cancer were independently associated with death., Interpretation: Our study demonstrates a high rate of nosocomial transmission of COVID-19, associated with increased mortality in both univariate and multivariate analysis in the cancer population, reinforcing the importance of treating patients with cancer in COVID-free units. We also validated that age and advanced cancer were negative predictive factors for COVID-19 severity in patients with cancer., Competing Interests: Conflict of interest statement BR and AE declare grant support from Astra Zeneca (grant number: N/A). Other authors have no conflict of interest to declare., (Copyright © 2020 Elsevier Ltd. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
42. Using Two Peptide Isotopologues as Internal Standards for the Streamlined Quantification of Low-Abundance Proteins by Immuno-MRM and Immuno-MALDI.
- Author
-
Ibrahim S, Froehlich BC, Aguilar-Mahecha A, Aloyz R, Poetz O, Basik M, Batist G, Zahedi RP, and Borchers CH
- Subjects
- Calibration, Cell Line, Tumor, Humans, Isotope Labeling, Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization standards, Colonic Neoplasms diagnosis, Enzyme-Linked Immunosorbent Assay standards, Peptides chemistry, Proteins analysis
- Abstract
Mass spectrometry (MS), particularly targeted proteomics, is increasingly being used for quantifying specific proteins and peptides in clinical specimens. The coupling of immuno-enrichment of proteotypic peptides with MS [e.g., immuno-multiple reaction monitoring (MRM) and immuno-matrix-assisted laser desorption ionization (MALDI)] enables the development of highly sensitive and specific assays for low-abundance signaling proteins. By incorporating stable isotope-labeled standards, these workflows allow the determination of endogenous protein concentrations. This is typically achieved through external calibration, often using surrogate matrices, which has inherent limitations for the analysis of clinical specimens as there are often substantial variations in the sample matrix, and sample amounts are typically limited. We have previously introduced the use of two peptide isotopologues for generating external calibration curves in plasma. Here, we present a two-point internal calibration (2-PIC) strategy using two isotopologues for immuno-MS assays and demonstrate its flexibility and robustness. Quantification of the tumor suppressor PTEN in Colo-205 cells by immuno-MRM and immuno-MALDI using 2-PIC and external calibration yielded very similar results (relative standard deviation between 2-PIC and external calibration: 4.9% for immuno-MRM; 1.1% for immuno-MALDI), without the need for a surrogate matrix or additional patient material for calibration, while concurrently reducing the instrument time and cost. Although our PTEN immuno-MRM and immuno-MALDI assays can be considered to be orthogonal as they utilized entirely different sample preparation and MS analysis workflows, targeted different PTEN peptides, and were performed in different laboratories, the endogenous Colo-205 PTEN levels determined with 2-PIC showed a good correlation ( r
2 = 0.9966) and good agreement (0.48 ± 0.01 and 0.29 ± 0.02 fmol/μg of total protein) between immuno-MRM and immuno-MALDI.- Published
- 2020
- Full Text
- View/download PDF
43. A Phase I, Dose Escalation, Single Dose Trial of Oral Attenuated Salmonella typhimurium Containing Human IL-2 in Patients With Metastatic Gastrointestinal Cancers.
- Author
-
Gniadek TJ, Augustin L, Schottel J, Leonard A, Saltzman D, Greeno E, and Batist G
- Subjects
- Biological Therapy adverse effects, Biomarkers, Combined Modality Therapy adverse effects, Combined Modality Therapy methods, Disease Management, Female, Gastrointestinal Neoplasms diagnosis, Gastrointestinal Neoplasms etiology, Humans, Immunotherapy methods, Interleukin-2 adverse effects, Lymphocytes immunology, Lymphocytes metabolism, Male, Neoplasm Metastasis, Neoplasm Staging, Palliative Care, Practice Guidelines as Topic, Biological Therapy methods, Gastrointestinal Neoplasms therapy, Interleukin-2 administration & dosage, Salmonella typhimurium immunology
- Abstract
Salmonella has been shown to preferentially colonize solid tumors. It is known that toxicity limits the systemic administration of immunomodulatory cytokines that have a significant anticancer effect. Therefore, we tested a unique cancer treatment strategy comprised of oral delivery of Saltikva, an attenuated strain of Salmonella typhimurium that contain the human gene for interleukin-2. In preclinical experimentation, a significant antitumor effect without toxicity was observed. A dose escalation, single dose, Phase I trial was conducted. Dose escalation (10 to 10) while monitoring for dose limiting toxicity and response was performed. Flow cytometry was conducted to determine the immunologic effect. In total 22 patients were administered Saltikva. Eight patients did not complete the trial. No toxicity or adverse events were observed. There was no survival advantage. Flow cytometry demonstrated an increase in circulating natural killer (NK) cells and NK-T cells when comparing the prestudy period. The results of this phase I dose escalation study show that oral attenuated S. typhimurium containing the human interleukin-2 gene caused no significant toxicities up to doses of 10 colony forming unit. There was no evidence of partial or complete response. All patients had progressive disease and eventually succumbed to their illness. Although no survival advantage was seen in this single dose study, the statistically significant increase in circulating NK and NK-T cell demonstrates an immunologic effect from this treatment regimen and suggest that a multiple dose study should be undertaken.
- Published
- 2020
- Full Text
- View/download PDF
44. Systematic Optimization of the iMALDI Workflow for the Robust and Straightforward Quantification of Signaling Proteins in Cancer Cells.
- Author
-
Froehlich BC, Popp R, Sobsey CA, Ibrahim S, LeBlanc AM, Mohammed Y, Aguilar-Mahecha A, Poetz O, Chen MX, Spatz A, Basik M, Batist G, Zahedi RP, and Borchers CH
- Subjects
- Cell Line, Tumor, Humans, Limit of Detection, Time Factors, Neoplasm Proteins metabolism, Signal Transduction, Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization methods, Workflow
- Abstract
Purpose: Immuno-MALDI (iMALDI) combines immuno-enrichment of biomarkers with MALDI-MS for fast, precise, and specific quantitation, making it a valuable tool for developing clinical assays. iMALDI assays are optimized for the PI3-kinase signaling pathway members phosphatase and tensin homolog (PTEN) and PI3-kinase catalytic subunit alpha (p110α), with regard to sensitivity, robustness, and throughput. A standardized template for developing future iMALDI assays, including automation protocols to streamline assay development and translation, is provided., Experimental Design: Conditions for tryptic digestion and immuno-enrichment (beads, bead:antibody ratios, incubation times, direct vs. indirect immuno-enrichment) are rigorously tested. Different strategies for calibration and data readout are compared., Results: Digestion using 1:2 protein:trypsin (wt:wt) for 1 h yielded high and consistent peptide recoveries. Direct immuno-enrichment (antibody-bead coupling prior to antigen-enrichment) yielded 30% higher peptide recovery with a 1 h shorter incubation time than indirect enrichment. Immuno-enrichment incubation overnight yielded 1.5-fold higher sensitivities than 1 h incubation. Quantitation of the endogenous target proteins is not affected by the complexity of the calibration matrix, further simplifying the workflow., Conclusions and Clinical Relevance: This optimized and automated workflow will facilitate the clinical translation of high-throughput sensitive iMALDI assays for quantifying cell-signaling proteins in individual tumor samples, thereby improving patient stratification for targeted treatment., (© 2020 The Authors. Proteomics - Clinical Applications published by Wiley-VCH GmbH.)
- Published
- 2020
- Full Text
- View/download PDF
45. Co-presence of human papillomaviruses and Epstein-Barr virus is linked with advanced tumor stage: a tissue microarray study in head and neck cancer patients.
- Author
-
Al-Thawadi H, Gupta I, Jabeen A, Skenderi F, Aboulkassim T, Yasmeen A, Malki MI, Batist G, Vranic S, and Al Moustafa AE
- Abstract
Background: Human papillomaviruses (HPVs) and Epstein-Barr virus (EBV), known oncoviruses, can be co-present and cooperate in the initiation and/or progression of human carcinomas, including head and neck. Based on this fact, we recently reported the prevalence of both HPVs and EBV in cervical and breast cancers., Methods: We herein explore for the first time the co-prevalence of high-risk HPVs and EBV in 98 head and neck (HN) squamous cell carcinoma (SCC) tissues from Bosnian patients using polymerase chain reaction (PCR) and immunohistochemistry (IHC) analysis, as well as tissue microarray methodology., Results: The majority of these cancer tissue cases were from the oral cavity (68%). We found that high-risk HPVs and EBV are co-present in 34.7% of the SCC samples; with a significant correlation between the various HPV types and EBV co-incidence (p = 0.03). Our data showed that 30.8% of oral SCCs are positive for E6 oncoprotein of high-risk HPVs and 44.6% are positive for LMP1 of EBV. The most commonly expressed HPVs in our HNSCC samples include HPV types 16, 18, 45 and 58. Additionally, 37.5% of oral SCCs are positive for both HPVs and EBV, with statistically significant association between high-risk HPV types and EBV (p < 0.05). More importantly, our data revealed that the co-presence of HPV and EBV is strongly correlated with advanced tumor stage (p = 0.035)., Conclusion: In this study we show that HPV and EBV oncoviruses are co-present in HNSCC, particularly in oral cancer, where they can cooperate in the initiation and/or progression of this cancer. Thus, further studies are necessary to elucidate the mechanism of this cooperation., Competing Interests: Competing interestsThe authors declare that they have no competing interests., (© The Author(s) 2020.)
- Published
- 2020
- Full Text
- View/download PDF
46. The Genomic Landscape of Intrinsic and Acquired Resistance to Cyclin-Dependent Kinase 4/6 Inhibitors in Patients with Hormone Receptor-Positive Metastatic Breast Cancer.
- Author
-
Wander SA, Cohen O, Gong X, Johnson GN, Buendia-Buendia JE, Lloyd MR, Kim D, Luo F, Mao P, Helvie K, Kowalski KJ, Nayar U, Waks AG, Parsons SH, Martinez R, Litchfield LM, Ye XS, Yu C, Jansen VM, Stille JR, Smith PS, Oakley GJ, Chu QS, Batist G, Hughes ME, Kremer JD, Garraway LA, Winer EP, Tolaney SM, Lin NU, Buchanan SG, and Wagle N
- Subjects
- Biopsy, Breast Neoplasms drug therapy, Breast Neoplasms pathology, Breast Neoplasms surgery, Cell Line, Tumor, Checkpoint Kinase 1, Female, Genomics, Humans, Proto-Oncogene Proteins c-akt, Proto-Oncogene Proteins p21(ras), Receptors, Steroid genetics, Retinoblastoma Binding Proteins, Ubiquitin-Protein Ligases, Exome Sequencing, Antineoplastic Agents therapeutic use, Breast Neoplasms genetics, Cell Cycle Proteins antagonists & inhibitors, Drug Resistance, Neoplasm genetics, Protein Kinase Inhibitors therapeutic use
- Abstract
Mechanisms driving resistance to cyclin-dependent kinase 4/6 inhibitors (CDK4/6i) in hormone receptor-positive (HR
+ ) breast cancer have not been clearly defined. Whole-exome sequencing of 59 tumors with CDK4/6i exposure revealed multiple candidate resistance mechanisms including RB1 loss, activating alterations in AKT1, RAS, AURKA, CCNE2, ERBB2 , and FGFR2 , and loss of estrogen receptor expression. In vitro experiments confirmed that these alterations conferred CDK4/6i resistance. Cancer cells cultured to resistance with CDK4/6i also acquired RB1, KRAS, AURKA , or CCNE2 alterations, which conferred sensitivity to AURKA, ERK, or CHEK1 inhibition. Three of these activating alterations-in AKT1, RAS , and AURKA -have not, to our knowledge, been previously demonstrated as mechanisms of resistance to CDK4/6i in breast cancer preclinically or in patient samples. Together, these eight mechanisms were present in 66% of resistant tumors profiled and may define therapeutic opportunities in patients. SIGNIFICANCE: We identified eight distinct mechanisms of resistance to CDK4/6i present in 66% of resistant tumors profiled. Most of these have a therapeutic strategy to overcome or prevent resistance in these tumors. Taken together, these findings have critical implications related to the potential utility of precision-based approaches to overcome resistance in many patients with HR+ metastatic breast cancer. This article is highlighted in the In This Issue feature, p. 1079 ., (©2020 American Association for Cancer Research.)- Published
- 2020
- Full Text
- View/download PDF
47. Evaluating the quantity, quality and size distribution of cell-free DNA by multiplex droplet digital PCR.
- Author
-
Alcaide M, Cheung M, Hillman J, Rassekh SR, Deyell RJ, Batist G, Karsan A, Wyatt AW, Johnson N, Scott DW, and Morin RD
- Subjects
- Biomarkers, Tumor genetics, Humans, Receptors, Odorant genetics, Cell-Free Nucleic Acids genetics, Polymerase Chain Reaction methods
- Abstract
Cell-free DNA (cfDNA) has become a comprehensive biomarker in the fields of non-invasive cancer detection and monitoring, organ transplantation, prenatal genetic testing and pathogen detection. While cfDNA samples can be obtained using a broad variety of approaches, there is an urgent need to standardize analytical tools aimed at assessing its basic properties. Typical methods to determine the yield and fragment size distribution of cfDNA samples are usually either blind to genomic DNA contamination or the presence of enzymatic inhibitors, which can confound and undermine downstream analyses. Here, we present a novel droplet digital PCR assay to identify suboptimal samples and aberrant cfDNA size distributions, the latter typically associated with high levels of circulating tumour DNA (ctDNA). Our assay was designed to promiscuously cross-amplify members of the human olfactory receptor (OR) gene family and includes a customizable diploid locus for the determination of absolute cfDNA concentrations. We demonstrate here the utility of our assay to estimate the yield and quality of cfDNA extracts and deduce fragment size distributions that correlate well with those inferred by capillary electrophoresis and high throughput sequencing. The assay described herein is a powerful tool to establish quality controls and stratify cfDNA samples based on presumed ctDNA levels, then facilitating the implementation of robust, cost-effective and standardized analytical workflows into clinical practice.
- Published
- 2020
- Full Text
- View/download PDF
48. Clinical impact of COVID-19 on patients with cancer (CCC19): a cohort study.
- Author
-
Kuderer NM, Choueiri TK, Shah DP, Shyr Y, Rubinstein SM, Rivera DR, Shete S, Hsu CY, Desai A, de Lima Lopes G Jr, Grivas P, Painter CA, Peters S, Thompson MA, Bakouny Z, Batist G, Bekaii-Saab T, Bilen MA, Bouganim N, Larroya MB, Castellano D, Del Prete SA, Doroshow DB, Egan PC, Elkrief A, Farmakiotis D, Flora D, Galsky MD, Glover MJ, Griffiths EA, Gulati AP, Gupta S, Hafez N, Halfdanarson TR, Hawley JE, Hsu E, Kasi A, Khaki AR, Lemmon CA, Lewis C, Logan B, Masters T, McKay RR, Mesa RA, Morgans AK, Mulcahy MF, Panagiotou OA, Peddi P, Pennell NA, Reynolds K, Rosen LR, Rosovsky R, Salazar M, Schmidt A, Shah SA, Shaya JA, Steinharter J, Stockerl-Goldstein KE, Subbiah S, Vinh DC, Wehbe FH, Weissmann LB, Wu JT, Wulff-Burchfield E, Xie Z, Yeh A, Yu PP, Zhou AY, Zubiri L, Mishra S, Lyman GH, Rini BI, and Warner JL
- Subjects
- Aged, Antiviral Agents therapeutic use, Azithromycin therapeutic use, Betacoronavirus, COVID-19, Cause of Death, Comorbidity, Coronavirus Infections drug therapy, Coronavirus Infections mortality, Databases, Factual, Female, Humans, Hydroxychloroquine therapeutic use, Male, Middle Aged, Neoplasms mortality, Neoplasms therapy, Pandemics, Pneumonia, Viral drug therapy, Pneumonia, Viral mortality, Prognosis, Risk Factors, SARS-CoV-2, COVID-19 Drug Treatment, Coronavirus Infections epidemiology, Neoplasms epidemiology, Pneumonia, Viral epidemiology
- Abstract
Background: Data on patients with COVID-19 who have cancer are lacking. Here we characterise the outcomes of a cohort of patients with cancer and COVID-19 and identify potential prognostic factors for mortality and severe illness., Methods: In this cohort study, we collected de-identified data on patients with active or previous malignancy, aged 18 years and older, with confirmed severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection from the USA, Canada, and Spain from the COVID-19 and Cancer Consortium (CCC19) database for whom baseline data were added between March 17 and April 16, 2020. We collected data on baseline clinical conditions, medications, cancer diagnosis and treatment, and COVID-19 disease course. The primary endpoint was all-cause mortality within 30 days of diagnosis of COVID-19. We assessed the association between the outcome and potential prognostic variables using logistic regression analyses, partially adjusted for age, sex, smoking status, and obesity. This study is registered with ClinicalTrials.gov, NCT04354701, and is ongoing., Findings: Of 1035 records entered into the CCC19 database during the study period, 928 patients met inclusion criteria for our analysis. Median age was 66 years (IQR 57-76), 279 (30%) were aged 75 years or older, and 468 (50%) patients were male. The most prevalent malignancies were breast (191 [21%]) and prostate (152 [16%]). 366 (39%) patients were on active anticancer treatment, and 396 (43%) had active (measurable) cancer. At analysis (May 7, 2020), 121 (13%) patients had died. In logistic regression analysis, independent factors associated with increased 30-day mortality, after partial adjustment, were: increased age (per 10 years; partially adjusted odds ratio 1·84, 95% CI 1·53-2·21), male sex (1·63, 1·07-2·48), smoking status (former smoker vs never smoked: 1·60, 1·03-2·47), number of comorbidities (two vs none: 4·50, 1·33-15·28), Eastern Cooperative Oncology Group performance status of 2 or higher (status of 2 vs 0 or 1: 3·89, 2·11-7·18), active cancer (progressing vs remission: 5·20, 2·77-9·77), and receipt of azithromycin plus hydroxychloroquine (vs treatment with neither: 2·93, 1·79-4·79; confounding by indication cannot be excluded). Compared with residence in the US-Northeast, residence in Canada (0·24, 0·07-0·84) or the US-Midwest (0·50, 0·28-0·90) were associated with decreased 30-day all-cause mortality. Race and ethnicity, obesity status, cancer type, type of anticancer therapy, and recent surgery were not associated with mortality., Interpretation: Among patients with cancer and COVID-19, 30-day all-cause mortality was high and associated with general risk factors and risk factors unique to patients with cancer. Longer follow-up is needed to better understand the effect of COVID-19 on outcomes in patients with cancer, including the ability to continue specific cancer treatments., Funding: American Cancer Society, National Institutes of Health, and Hope Foundation for Cancer Research., (Copyright © 2020 Elsevier Ltd. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
49. Tofacitinib for Refractory Immune-Related Colitis from PD-1 Therapy.
- Author
-
Esfahani K, Hudson M, and Batist G
- Subjects
- Humans, Piperidines, Programmed Cell Death 1 Receptor, Pyrimidines, Pyrroles, Colitis, Colitis, Ulcerative
- Published
- 2020
- Full Text
- View/download PDF
50. Targeted and Untargeted Proteomics Approaches in Biomarker Development.
- Author
-
Sobsey CA, Ibrahim S, Richard VR, Gaspar V, Mitsa G, Lacasse V, Zahedi RP, Batist G, and Borchers CH
- Subjects
- Biomarkers metabolism, Biomedical Research, Hemoglobinopathies blood, Hemoglobinopathies diagnosis, Humans, Immunoassay methods, Prostate-Specific Antigen analysis, Protein Isoforms analysis, Proteomics trends, Reproducibility of Results, Biomarkers analysis, Mass Spectrometry methods, Proteins analysis, Proteomics methods
- Abstract
An enormous amount of research effort has been devoted to biomarker discovery and validation. With the completion of the human genome, proteomics is now playing an increasing role in this search for new and better biomarkers. Here, what leads to successful biomarker development is reviewed and how these features may be applied in the context of proteomic biomarker research is considered. The "fit-for-purpose" approach to biomarker development suggests that untargeted proteomic approaches may be better suited for early stages of biomarker discovery, while targeted approaches are preferred for validation and implementation. A systematic screening of published biomarker articles using MS-based proteomics reveals that while both targeted and untargeted technologies are used in proteomic biomarker development, most researchers do not combine these approaches. i) The reasons for this discrepancy, (ii) how proteomic technologies can overcome technical challenges that seem to limit their translation into the clinic, and (iii) how MS can improve, complement, or replace existing clinically important assays in the future are discussed., (© 2019 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim.)
- Published
- 2020
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.