9 results on '"Joshua K. Sabari"'
Search Results
2. Dynamic Management of Lung Cancer Care During Surging COVID-19
- Author
-
Annie Wang, Stephanie H. Chang, Eric J. Kim, Jamie L. Bessich, Joshua K. Sabari, Benjamin Cooper, Travis C. Geraci, and Robert J. Cerfolio
- Subjects
lung cancer ,thoracic surgery ,COVID-19 ,radiation oncology ,screening ,Surgery ,RD1-811 - Abstract
Management of patients with lung cancer continues to be challenging during the COVID-19 pandemic, due to the increased risk of complications in this subset of patients. During the COVID-19 surge in New York City, New York University Langone Health adopted triage strategies to help with care for lung cancer patients, with good surgical outcomes and no transmission of COVID-19 to patients or healthcare workers. Here, we will review current recommendations regarding screening and management of lung cancer patients during both a non-surge phase and surge phase of COVID-19.
- Published
- 2021
- Full Text
- View/download PDF
3. Next-generation sequencing based detection of germline and somatic alterations in a patient with four metachronous primary tumors
- Author
-
Madhuri Martin, Joshua K. Sabari, Gulisa Turashvili, Darragh F. Halpenny, Hira Rizvi, Natalie Shapnik, and Vicky Makker
- Subjects
Gynecology and obstetrics ,RG1-991 ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Introduction: Multiple primary tumors (MPTs) are defined as two or more separate synchronous or metachronous neoplasms occurring in different sites in the same individual. These tumors differ in histology, as well as primary sites from which they arise. Risk factors associated with the occurrence of MPTs include germline alterations, exposure to prior cancer therapies, occupational hazards, and lifestyle and behavioral influences. Case report: We present a case of a patient who was diagnosed with four metachronous primary tumors. In 2013, she was diagnosed with serous proliferations associated with psammomatous bodies of primary peritoneal origin (pT3NxM0). This was followed by invasive ductal carcinoma of the breast (stage pT2N0Mx, histological grade III/III) in 2014, melanoma (stage pT2bNxMx) in 2016 that further advanced to the lung and brain in 2017, and a low-grade lung carcinoid in 2017. To better understand the biology of this patient's MPTs, we performed next-generation sequencing (NGS) to assess for both somatic and germline alterations. The treatment course for this patient aims to target the tumor with the strongest prognostic value, namely her malignant melanoma, and has contributed favorably to the overall survival of this patient. Conclusion: We report the clinical and genomic landscape of a patient with MPTs who had no identifiable unique somatic or germline mutations to explain her predilection to cancer. The treatment course and overall prognosis for this patient is important for understanding future cases with unrelated, metachronous MPTs, the occurrence of which cannot always be explained by underlying genetic mechanisms. Keywords: Multiple primary tumors, Next-generation sequencing, Tumor mutational burden, Immunotherapy
- Published
- 2018
- Full Text
- View/download PDF
4. Immune checkpoint inhibitors for patients with advanced lung cancer and oncogenic driver alterations: results from the IMMUNOTARGET registry
- Author
-
Alexis B. Cortot, Sébastien Couraud, M. van den Heuvel, Julie Milia, Benjamin Besse, S-H.I. Ou, Sacha I. Rothschild, Mickaël Lattuca-Truc, Laura Mezquita, Joel W. Neal, Rafael Rosell, Amélie Lusque, R.D. Schouten, Terry L. Ng, Martin Früh, Marcel Wiesweg, Benjamin Solomon, D.R. Camidge, Nir Peled, C. Mascaux, Gérard Zalcman, Alexander Drilon, Alessandra Curioni-Fontecedro, Heather A. Wakelee, Alex Martinez-Marti, Paolo Bironzo, Julien Mazieres, Sanjay Popat, Valérie Gounant, Laurent Mhanna, Remi Veillon, Viola W. Zhu, Oliver Gautschi, Fabrice Barlesi, Joshua K. Sabari, Silvia Novello, A. Thai, Joachim Diebold, Rangueil-Larrey University Hospital, Université Toulouse III - Paul Sabatier (UT3), Université Fédérale Toulouse Midi-Pyrénées-Université Fédérale Toulouse Midi-Pyrénées, Centre de Recherche en Cancérologie de Marseille (CRCM), Aix Marseille Université (AMU)-Institut Paoli-Calmettes, Fédération nationale des Centres de lutte contre le Cancer (FNCLCC)-Fédération nationale des Centres de lutte contre le Cancer (FNCLCC)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Service de pneumologie [Centre Hospitalier Lyon Sud - HCL], Centre Hospitalier Lyon Sud [CHU - HCL] (CHLS), Hospices Civils de Lyon (HCL)-Hospices Civils de Lyon (HCL), CHU Tenon [AP-HP], Sorbonne Université (SU)-Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP), Intergroupe Francophone de Cancérologie Thoracique [Paris] (IFCT), Intergroupe Francophone de Cancérologie thoracique, Service d'oncologie multidisciplinaire innovations thérapeutiques [Hôpital Nord - APHM], Assistance Publique - Hôpitaux de Marseille (APHM)- Hôpital Nord [CHU - APHM], Centre Hospitalier Universitaire de Toulouse (CHU Toulouse), and Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Sorbonne Université (SU)
- Subjects
0301 basic medicine ,Oncology ,Male ,Lung Neoplasms ,[SDV]Life Sciences [q-bio] ,Programmed Cell Death 1 Receptor ,Medizin ,medicine.disease_cause ,B7-H1 Antigen ,Antineoplastic Agents, Immunological ,0302 clinical medicine ,Carcinoma, Non-Small-Cell Lung ,Antineoplastic Combined Chemotherapy Protocols ,80 and over ,Registries ,Non-Small-Cell Lung ,Lung ,Cancer ,Aged, 80 and over ,Tumor ,Hematology ,Middle Aged ,Chemotherapy regimen ,Progression-Free Survival ,3. Good health ,Immunotherapy-lung cancer-oncogenic addiction ,Editorial Commentary ,Immunological ,Response Evaluation Criteria in Solid Tumors ,5.1 Pharmaceuticals ,030220 oncology & carcinogenesis ,Adenocarcinoma ,Female ,KRAS ,immunotherapy ,Development of treatments and therapeutic interventions ,hormones, hormone substitutes, and hormone antagonists ,Adult ,medicine.medical_specialty ,Clinical Trials and Supportive Activities ,Oncology and Carcinogenesis ,Context (language use) ,Antineoplastic Agents ,and over ,[SDV.BC]Life Sciences [q-bio]/Cellular Biology ,Rare cancers Radboud Institute for Molecular Life Sciences [Radboudumc 9] ,03 medical and health sciences ,Clinical Research ,Internal medicine ,Biomarkers, Tumor ,medicine ,ROS1 ,Humans ,Progression-free survival ,Oncology & Carcinogenesis ,Lung cancer ,oncogenic addiction ,neoplasms ,Retrospective Studies ,Aged ,Thoracic tumor ,business.industry ,Carcinoma ,Oncogenes ,medicine.disease ,respiratory tract diseases ,lung cancer ,030104 developmental biology ,Good Health and Well Being ,Mutation ,business ,Biomarkers - Abstract
Contains fulltext : 215810.pdf (Publisher’s version ) (Closed access) BACKGROUND: Anti-PD1/PD-L1 directed immune checkpoint inhibitors (ICI) are widely used to treat patients with advanced non-small-cell lung cancer (NSCLC). The activity of ICI across NSCLC harboring oncogenic alterations is poorly characterized. The aim of our study was to address the efficacy of ICI in the context of oncogenic addiction. PATIENTS AND METHODS: We conducted a retrospective study for patients receiving ICI monotherapy for advanced NSCLC with at least one oncogenic driver alteration. Anonymized data were evaluated for clinicopathologic characteristics and outcomes for ICI therapy: best response (RECIST 1.1), progression-free survival (PFS), and overall survival (OS) from ICI initiation. The primary end point was PFS under ICI. Secondary end points were best response (RECIST 1.1) and OS from ICI initiation. RESULTS: We studied 551 patients treated in 24 centers from 10 countries. The molecular alterations involved KRAS (n = 271), EGFR (n = 125), BRAF (n = 43), MET (n = 36), HER2 (n = 29), ALK (n = 23), RET (n = 16), ROS1 (n = 7), and multiple drivers (n = 1). Median age was 60 years, gender ratio was 1 : 1, never/former/current smokers were 28%/51%/21%, respectively, and the majority of tumors were adenocarcinoma. The objective response rate by driver alteration was: KRAS = 26%, BRAF = 24%, ROS1 = 17%, MET = 16%, EGFR = 12%, HER2 = 7%, RET = 6%, and ALK = 0%. In the entire cohort, median PFS was 2.8 months, OS 13.3 months, and the best response rate 19%. In a subgroup analysis, median PFS (in months) was 2.1 for EGFR, 3.2 for KRAS, 2.5 for ALK, 3.1 for BRAF, 2.5 for HER2, 2.1 for RET, and 3.4 for MET. In certain subgroups, PFS was positively associated with PD-L1 expression (KRAS, EGFR) and with smoking status (BRAF, HER2). CONCLUSIONS: : ICI induced regression in some tumors with actionable driver alterations, but clinical activity was lower compared with the KRAS group and the lack of response in the ALK group was notable. Patients with actionable tumor alterations should receive targeted therapies and chemotherapy before considering immunotherapy as a single agent.
- Published
- 2019
- Full Text
- View/download PDF
5. Comprehensive Molecular and Clinicopathologic Analysis of 200 Pulmonary Invasive Mucinous Adenocarcinomas Identifies Distinct Characteristics of Molecular Subtypes
- Author
-
Brian Houck-Loomis, Joshua K. Sabari, Ryma Benayed, Laetitia Borsu, Kyuichi Kadota, Helen Won, Prasad S. Adusumilli, Alison M. Schram, Maria E. Arcila, Jason C. Chang, Vasilisa A. Rudneva, Natasha Rekhtman, Sarah Teed, Valerie W. Rusch, Christina Falcon, Khedoudja Nafa, Marc Ladanyi, Patrice Desmeules, Michael Offin, Fanli Meng, Bob T. Li, William D. Travis, Alexander Drilon, David N Brown, Sharon Amir, and Joseph Montecalvo
- Subjects
0301 basic medicine ,Adult ,Male ,Cancer Research ,Lung Neoplasms ,medicine.medical_treatment ,Biology ,medicine.disease_cause ,Mutually exclusive events ,Article ,Targeted therapy ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Humans ,Neoplasm Invasiveness ,Gene ,Genotyping ,Aged ,Aged, 80 and over ,High prevalence ,Middle Aged ,medicine.disease ,Adenocarcinoma, Mucinous ,Mucinous Adenocarcinomas ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Mutation ,Cancer research ,Adenocarcinoma ,Female ,KRAS - Abstract
Purpose:Invasive mucinous adenocarcinoma (IMA) is a unique subtype of lung adenocarcinoma, characterized genomically by frequent KRAS mutations or specific gene fusions, most commonly involving NRG1. Comprehensive analysis of a large series of IMAs using broad DNA- and RNA-sequencing methods is still lacking, and it remains unclear whether molecular subtypes of IMA differ clinicopathologically.Experimental Design:A total of 200 IMAs were analyzed by 410-gene DNA next-generation sequencing (MSK-IMPACT; n = 136) or hotspot 8-oncogene genotyping (n = 64). Driver-negative cases were further analyzed by 62-gene RNA sequencing (MSK-Fusion) and those lacking fusions were further tested by whole-exome sequencing and whole-transcriptome sequencing (WTS).Results:Combined MSK-IMPACT and MSK-Fusion testing identified mutually exclusive driver alterations in 96% of IMAs, including KRAS mutations (76%), NRG1 fusions (7%), ERBB2 alterations (6%), and other less common events. In addition, WTS identified a novel NRG2 fusion (F11R–NRG2). Overall, targetable gene fusions were identified in 51% of KRAS wild-type IMAs, leading to durable responses to targeted therapy in some patients. Compared with KRAS-mutant IMAs, NRG1-rearranged tumors exhibited several more aggressive characteristics, including worse recurrence-free survival (P < 0.0001).Conclusions:This is the largest molecular study of IMAs to date, where we demonstrate the presence of a major oncogenic driver in nearly all cases. This study is the first to document more aggressive characteristics of NRG1-rearranged IMAs, ERBB2 as the third most common alteration, and a novel NRG2 fusion in these tumors. Comprehensive molecular testing of KRAS wild-type IMAs that includes fusion testing is essential, given the high prevalence of alterations with established and investigational targeted therapies in this subset.
- Published
- 2021
6. Next-generation sequencing based detection of germline and somatic alterations in a patient with four metachronous primary tumors
- Author
-
Hira Rizvi, Darragh Halpenny, Joshua K. Sabari, Madhuri Martin, Vicky Makker, Natalie Shapnik, and Gulisa Turashvili
- Subjects
0301 basic medicine ,Oncology ,medicine.medical_specialty ,Somatic cell ,lcsh:Gynecology and obstetrics ,lcsh:RC254-282 ,Germline ,03 medical and health sciences ,0302 clinical medicine ,Germline mutation ,Internal medicine ,medicine ,Stage (cooking) ,lcsh:RG1-991 ,business.industry ,Melanoma ,Obstetrics and Gynecology ,Cancer ,Histology ,medicine.disease ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Serous fluid ,030104 developmental biology ,030220 oncology & carcinogenesis ,business - Abstract
Introduction: Multiple primary tumors (MPTs) are defined as two or more separate synchronous or metachronous neoplasms occurring in different sites in the same individual. These tumors differ in histology, as well as primary sites from which they arise. Risk factors associated with the occurrence of MPTs include germline alterations, exposure to prior cancer therapies, occupational hazards, and lifestyle and behavioral influences. Case report: We present a case of a patient who was diagnosed with four metachronous primary tumors. In 2013, she was diagnosed with serous proliferations associated with psammomatous bodies of primary peritoneal origin (pT3NxM0). This was followed by invasive ductal carcinoma of the breast (stage pT2N0Mx, histological grade III/III) in 2014, melanoma (stage pT2bNxMx) in 2016 that further advanced to the lung and brain in 2017, and a low-grade lung carcinoid in 2017. To better understand the biology of this patient's MPTs, we performed next-generation sequencing (NGS) to assess for both somatic and germline alterations. The treatment course for this patient aims to target the tumor with the strongest prognostic value, namely her malignant melanoma, and has contributed favorably to the overall survival of this patient. Conclusion: We report the clinical and genomic landscape of a patient with MPTs who had no identifiable unique somatic or germline mutations to explain her predilection to cancer. The treatment course and overall prognosis for this patient is important for understanding future cases with unrelated, metachronous MPTs, the occurrence of which cannot always be explained by underlying genetic mechanisms. Keywords: Multiple primary tumors, Next-generation sequencing, Tumor mutational burden, Immunotherapy
- Published
- 2018
7. Immunophenotype and Response to Immunotherapy of RET-Rearranged Lung Cancers
- Author
-
Dazhi Liu, Joshua K. Sabari, Bob T. Li, Charles M. Rudin, Robin Guo, Natasha Rekhtman, Gregory J. Riely, Maria E. Arcila, Stephanie L. Wu, Marc Ladanyi, Ai Ni, Ryma Benayed, Terry Pak, Mark G. Kris, Joseph Montecalvo, Alexander Drilon, Michael Offin, Josiah D. Land, Larry W Buie, Darragh Halpenny, and Matthew D. Hellmann
- Subjects
Oncology ,0303 health sciences ,Cancer Research ,medicine.medical_specialty ,Chemotherapy ,Lung ,business.industry ,medicine.medical_treatment ,Immunotherapy ,Disease ,Immune checkpoint ,Article ,3. Good health ,Blockade ,Targeted therapy ,03 medical and health sciences ,0302 clinical medicine ,Immunophenotyping ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Internal medicine ,medicine ,business ,030304 developmental biology - Abstract
RET rearrangements are identified in 1% to 2% of non-small-cell lung cancers (NSCLCs).1,2 In patients with advanced, RET-rearranged lung cancers, systemic therapy can be highly active. We demonstrated previously that pemetrexed-based chemotherapy can achieve an objective response rate of 45% and a median progression-free survival (PFS) of 19 months.3 Furthermore, the activity of targeted therapy has improved dramatically with the introduction of selective RET inhibitors to the clinic. In early-phase testing, objective response rates with LOXO-2924 and BLU-6675 are 68% (26 of 38) and 50% (seven of 14), respectively. These outcomes exceed the modest activity observed previously with multikinase inhibitors such as cabozantinib6 and vandetanib.7 In contrast, the activity of immunotherapy in RET-rearranged lung cancers has not been well characterized. This represents a clear unmet need, given that all prior regulatory approvals of immune checkpoint inhibitors, either alone or in combination with chemotherapy, and in stage III or IV disease, have technically included patients with RET-rearranged lung cancers.8,9 Furthermore, although increasing levels of programmed death-ligand 1 (PD-L1) expression and high tumor mutational burden (TMB) have been associated with benefit from immune checkpoint blockade,10 the immunophenotype of RET-rearranged lung cancers and the role of PD-L1 and TMB status in relation to benefit with immunotherapy remain poorly described. We set out to characterize these factors.
- Published
- 2019
8. Impact of Baseline Steroids on Efficacy of Programmed Cell Death-1 and Programmed Death-Ligand 1 Blockade in Patients With Non-Small-Cell Lung Cancer
- Author
-
David Planchard, Niamh Long, W. Victoria Lai, Andrew J. Plodkowski, Kathryn C. Arbour, Matthew D. Hellmann, Roberto Ferrara, Benjamin Besse, Edouard Auclin, Laura Mezquita, Joshua K. Sabari, Darragh Halpenny, Jamie E. Chaft, Lizza E.L. Hendriks, Gregory J. Riely, Caroline Caramella, Hira Rizvi, Gala Martínez-Bernal, Andy Ni, RS: GROW - R3 - Innovative Cancer Diagnostics & Therapy, Pulmonologie, Promovendi ODB, and MUMC+: MA Med Staf Spec Longziekten (9)
- Subjects
0301 basic medicine ,Adult ,Male ,Cancer Research ,Lung Neoplasms ,Cell ,Programmed Cell Death 1 Receptor ,Pembrolizumab ,FATIGUE ,B7-H1 Antigen ,03 medical and health sciences ,DOUBLE-BLIND ,0302 clinical medicine ,Antineoplastic Agents, Immunological ,Adrenal Cortex Hormones ,Carcinoma, Non-Small-Cell Lung ,Carcinoma ,medicine ,Humans ,Drug Interactions ,Adverse effect ,Lung cancer ,PEMBROLIZUMAB ,Dexamethasone ,DYSPNEA ,Aged ,DOCETAXEL ,business.industry ,Middle Aged ,medicine.disease ,OPEN-LABEL ,Blockade ,DEXAMETHASONE ,030104 developmental biology ,medicine.anatomical_structure ,Treatment Outcome ,Oncology ,Docetaxel ,030220 oncology & carcinogenesis ,Cancer research ,T-CELLS ,Female ,ADVERSE EVENTS ,business ,medicine.drug - Abstract
Purpose Treatment with programmed cell death-1 or programmed death ligand 1 (PD-(L)1) inhibitors is now standard therapy for patients with lung cancer. The immunosuppressive effect of corticosteroids may reduce efficacy of PD-(L)1 blockade. On-treatment corticosteroids for treatment of immune-related adverse events do not seem to affect efficacy, but the potential impact of baseline corticosteroids at the time of treatment initiation is unknown. Clinical trials typically excluded patients who received baseline corticosteroids, which led us to use real-world data to examine the effect of corticosteroids at treatment initiation. Methods We identified patients who were PD-(L)1–naïve with advanced non–small-cell lung cancer from two institutions—Memorial Sloan Kettering Cancer Center and Gustave Roussy Cancer Center—who were treated with single-agent PD-(L)1 blockade. Clinical and pharmacy records were reviewed to identify corticosteroid use at the time of beginning anti–PD-(L)1 therapy. We performed multivariable analyses using Cox proportional hazards regression model and logistic regression. Results Ninety (14%) of 640 patients treated with single-agent PD-(L)1 blockade received corticosteroids of ≥ 10 mg of prednisone equivalent daily at the start of the PD-(L)1 blockade. Common indications for corticosteroids were dyspnea (33%), fatigue (21%), and brain metastases (19%). In both independent cohorts, Memorial Sloan Kettering Cancer Center (n = 455) and Gustave Roussy Cancer Center (n = 185), baseline corticosteroids were associated with decreased overall response rate, progression-free survival, and overall survival with PD-(L)1 blockade. In a multivariable analysis of the pooled population, adjusting for smoking history, performance status, and history of brain metastases, baseline corticosteroids remained significantly associated with decreased progression-free survival (hazard ratio, 1.3; P = .03), and overall survival (hazard ratio, 1.7; P < .001). Conclusion Baseline corticosteroid use of ≥ 10 mg of prednisone equivalent was associated with poorer outcome in patients with non–small-cell lung cancer who were treated with PD-(L)1 blockade. Prudent use of corticosteroids at the time of initiating PD-(L)1 blockade is recommended.
- Published
- 2018
9. Impact of Baseline Steroids on Efficacy of Programmed Cell Death-1 and Programmed Death-Ligand 1 Blockade in Patients With Non-Small-Cell Lung Cancer.
- Author
-
Arbour KC, Mezquita L, Long N, Rizvi H, Auclin E, Ni A, Martínez-Bernal G, Ferrara R, Lai WV, Hendriks LEL, Sabari JK, Caramella C, Plodkowski AJ, Halpenny D, Chaft JE, Planchard D, Riely GJ, Besse B, and Hellmann MD
- Subjects
- Adult, Aged, B7-H1 Antigen antagonists & inhibitors, Female, Humans, Male, Middle Aged, Programmed Cell Death 1 Receptor antagonists & inhibitors, Treatment Outcome, Adrenal Cortex Hormones therapeutic use, Antineoplastic Agents, Immunological therapeutic use, Carcinoma, Non-Small-Cell Lung drug therapy, Drug Interactions, Lung Neoplasms drug therapy
- Abstract
Purpose: Treatment with programmed cell death-1 or programmed death ligand 1 (PD-(L)1) inhibitors is now standard therapy for patients with lung cancer. The immunosuppressive effect of corticosteroids may reduce efficacy of PD-(L)1 blockade. On-treatment corticosteroids for treatment of immune-related adverse events do not seem to affect efficacy, but the potential impact of baseline corticosteroids at the time of treatment initiation is unknown. Clinical trials typically excluded patients who received baseline corticosteroids, which led us to use real-world data to examine the effect of corticosteroids at treatment initiation., Methods: We identified patients who were PD-(L)1-naïve with advanced non-small-cell lung cancer from two institutions-Memorial Sloan Kettering Cancer Center and Gustave Roussy Cancer Center-who were treated with single-agent PD-(L)1 blockade. Clinical and pharmacy records were reviewed to identify corticosteroid use at the time of beginning anti-PD-(L)1 therapy. We performed multivariable analyses using Cox proportional hazards regression model and logistic regression., Results: Ninety (14%) of 640 patients treated with single-agent PD-(L)1 blockade received corticosteroids of ≥ 10 mg of prednisone equivalent daily at the start of the PD-(L)1 blockade. Common indications for corticosteroids were dyspnea (33%), fatigue (21%), and brain metastases (19%). In both independent cohorts, Memorial Sloan Kettering Cancer Center (n = 455) and Gustave Roussy Cancer Center (n = 185), baseline corticosteroids were associated with decreased overall response rate, progression-free survival, and overall survival with PD-(L)1 blockade. In a multivariable analysis of the pooled population, adjusting for smoking history, performance status, and history of brain metastases, baseline corticosteroids remained significantly associated with decreased progression-free survival (hazard ratio, 1.3; P = .03), and overall survival (hazard ratio, 1.7; P < .001)., Conclusion: Baseline corticosteroid use of ≥ 10 mg of prednisone equivalent was associated with poorer outcome in patients with non-small-cell lung cancer who were treated with PD-(L)1 blockade. Prudent use of corticosteroids at the time of initiating PD-(L)1 blockade is recommended.
- Published
- 2018
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.