7 results on '"Jose V. Forero"'
Search Results
2. ICOS and OX40 Signaling Optimization Improves CAR-T Cell Persistence in Preclinical Models
- Author
-
Eider Moreno, Pedro Franco-Fuquen, Jose V. Forero, Juan Esteban Garcia Robledo, Natalie Booth, and Januario E. Castro
- Subjects
Immunology ,Cell Biology ,Hematology ,Biochemistry - Published
- 2022
3. Ibrutinib plus Obinutuzumab as Frontline Therapy for Chronic Lymphocytic Leukemia Is Associated with a Lower Rate of Infusion-Related Reactions and with Sustained Remissions after Ibrutinib Discontinuation: A Single-Arm, Open-Label, Phase 1b/2 Clinical Trial NCT0231576
- Author
-
Juliana Velez-Lujan, Jose V. Forero, Paula A. Lengerke-Diaz, Carlos I. Amaya-Chanaga, Alaina Heinen, Juan Esteban Garcia-Robledo, Colin McCarthy, Michale Y. Choi, Eider F. Moreno-Cortes, Chaja Jacobs, Thomas J. Kipps, Januario E. Castro, and Matutes, Estella M
- Subjects
Oncology ,medicine.medical_specialty ,Article Subject ,Lymphoma ,Chronic lymphocytic leukemia ,Rate of infusion ,Clinical Trials and Supportive Activities ,chemistry.chemical_compound ,Rare Diseases ,Obinutuzumab ,Clinical Research ,Internal medicine ,medicine ,Diseases of the blood and blood-forming organs ,6.2 Cellular and gene therapies ,Cancer ,business.industry ,Evaluation of treatments and therapeutic interventions ,Hematology ,medicine.disease ,Discontinuation ,Clinical trial ,chemistry ,Ibrutinib ,6.1 Pharmaceuticals ,Open label ,RC633-647.5 ,business ,Research Article - Abstract
Ibrutinib-based therapies are costly and require continuous administration. We hypothesized combining BTK inhibition with anti-CD20 monoclonal antibodies would yield deep remissions allowing discontinuation. We enrolled 32 therapy-naïve CLL patients to receive ibrutinib plus obinutuzumab, followed by single-agent ibrutinib. Patients could discontinue ibrutinib after 36 months with sustained complete response (CR). We evaluated treatment safety, efficacy, and outcomes after ibrutinib discontinuation. The overall response rate was 100%, 28% achieved a CR, and 12.5% achieved bone marrow undetectable minimal residual disease. At a three-year median follow-up, 91% remain in remission with 100% overall survival. Five patients in sustained CR stopped ibrutinib and have not progressed. Eight non-CR patients discontinued for other reasons, with only two progressing. The treatment was safe, with a lower IRR rate. All patients responded to treatment with longer time-to-progression after discontinuation of ibrutinib. Our data support the evaluation of ibrutinib discontinuation strategies in more extensive clinical trials (https://Clinicaltrials.gov Identifier https://clinicaltrials.gov/ct2/show/NCT02315768).
- Published
- 2022
4. Preclinical NK Cell Platform for CAR Directed Therapies: Functional and Phenotypic Comparison Using a Rechallenge Cytotoxicity Assay
- Author
-
Jose V. Forero, Eider F. Moreno Cortes, Natalie Booth, Januario E. Castro, and Juan Esteban Garcia Robledo
- Subjects
medicine.anatomical_structure ,Immunology ,Cell ,Cancer research ,medicine ,Cell Biology ,Hematology ,Biology ,Cytotoxicity ,Biochemistry ,Phenotype - Abstract
Introduction: CAR T-cell therapy has revolutionized the treatment of patients with relapsed/refractory (R/R) acute leukemia, NHL, and multiple myeloma. However, there are still areas of improvement in their clinical activity, source of the effector cells, prevention, and management of adverse events that require particular attention. Because of those reasons, NK cells appear as a viable effector cell alternative that can help address these challenges. NK cells offer a profile of activation, expansion, persistence, and cytotoxicity that is different from T cells and, when modified to bear CAR constructs, may provide significant advantages. However, the preclinical development of NK-CARs is challenging mainly because of the difficulty of generating large quantities of cells for testing and well-established pathways for CAR optimization before in vivo evaluation. Therefore, we developed a CAR optimization platform using the NK-92 cell line. NK-92 cells conserve their cytotoxic ability and can be easily expanded in vitro and used for functional and phenotypical evaluations of novel CAR-NK constructs. Here we present a rechallenge cytotoxic assay that mimics repetitive in vivo effector interactions with the target cells and its use for optimization, comparison, and development of NK-based cellular therapies. Methods: We generated lentivirus transduced CD19 CARs (FMC63-41BB-z) using T cells from healthy donors and NK-92 cells for comparison.T cells were expanded for 12 days, and a 41.9% CAR+ expression was achieved (CART19). Transduced NK-92 cells were sorted by FACS to obtain a population of 98.3 % CAR+ cells (CARNK19) and subsequently expanded for 12 days. JeKo-1 cells were used as CD19+ targets and BxPC3 cells as CD19 neg control (both cell types were GFP-Luc-PuroR). We developed a Luciferase-based rechallenge cytotoxicity assay. For this, we diluted the effector to target (E/T) ratio to obtain a logarithmic trendline of the cells' cytotoxicity. E/T ratio to get viability of 50% (IC50) measured at 4h (for CARNK19) and 24h (for CART19) was used as a proxy of the product's potency. Both CAR Immune Effector Cells (IECs) were co-cultured with their targets at an E/T ratio to obtain 70% cytotoxicity. After 24 hours with the target, we estimated the remaining IEC amount in the culture using GFP exclusion in flow analysis (IEC cells/mL = total cells/mL x GFP neg%). We repeated the plating of E/T ratio dilutions to perform daily IC50 curves using this rechallenge strategy for a total of 5 days. CAR and PD1 expression were measured on Day 0 and Day 5 by flow cytometry. Results: CART19 showed a higher IC50 than CARNK19 at baseline, 1.7 vs. 0.19 (Figure 1A). The IC50 trend of both IECs over time showed an uptrend that suggests progressive functional exhaustion (Figure 1B). At 5 days of rechallenge, it was 29 times higher in T cells than in NK-92 (12.07 vs. 0.42) and with a slope 265 times higher (10.6 vs. 0.04). Furthermore, we observed that when comparing the levels of CAR expression on Day 0 vs. Day 5, CART19 showed a decrease in CAR expression that was not present in CARNK19 (41.9 to 10.9% vs. 98.3 to 95.5%) (Figure 1C). In addition, there was a higher increase in PD1 expression in CART19 cells than CARNK19 cells from Day 0 to Day 5 of the in vitro rechallenge (9.9 to 46.8% vs. 0.88 to 8.88%) (Figure 1D). Conclusion: Our data shows the use of NK-92 cells as a tool for optimization and preclinical development of NK cell-based cellular therapies. We demonstrated that it is feasible to set up repetitive cytotoxic challenges that mimic closer in vivo E/T engagement. Moreover, using the cytotoxic IC50 calculated with this platform, we show increased cytotoxicity, less functional exhaustion, and less expression of PD1 in CARNK19 than in its T cell counterpart. Overall, the NK-92 rechallenge cytotoxicity assay platform constitutes a helpful tool for research, development, and optimization of cellular therapies based on NK cell effector function. Figure 1 Figure 1. Disclosures No relevant conflicts of interest to declare.
- Published
- 2021
5. Optimization of Third Generation Chimeric Antigen Receptor T Cells Targeting ROR1 for Hematological Malignancies
- Author
-
Juan Esteban Garcia Robledo, Jose V. Forero, Natalie Booth, Januario E. Castro, and Eider F. Moreno Cortes
- Subjects
Immunology ,ROR1 ,Cancer research ,Cell Biology ,Hematology ,Biology ,Biochemistry ,Third generation ,Chimeric antigen receptor - Abstract
Background: Chimeric Antigen Receptor (CAR) T cell therapy is arguably one of the most significant breakthroughs in cancer treatment. There are currently five FDA-approved products that are commercially available. However, despite their success, these CAR T-cell therapies cannot induce long-term durable responses in approximately 50% of leukemia or lymphoma-treated patients. Similarly, the results of CAR T-cells in solid tumors have been somewhat disappointing. Therefore, there is an urgent need to design and develop novel CAR T cells with improved efficacy in hematologic malignancies and solid tumors. ROR1 is a carcinoembryonic antigen expressed in different cancers and is associated with tumor stemness, proliferation, metastatic transformation, and treatment resistance. In this project, we optimize an anti-ROR1 CAR using a humanized single-chain variable fragment (scFv) with second (2G) or third-generation (3G) costimulatory domains. Methods: Several optimization steps in silico were performed using a selected scFv binding domain that targets ROR1. Those included codon optimizations, positional arrangement of heavy-light chains, evaluation of the ideal length of linkers based on tridimensional modeling of the docking between the antibody-like paratope with the target antigen (Figure 1A). After this initial scFv optimization process, we constructed a lentiviral vector that encodes CARs using the selected scFv linked to a transmembrane domain CD28 and different signaling endodomains for 2G and 3G variants (CD28, 41BB, ICOS, OX40), each linked to the T cell receptor CD3z domain. The cytotoxic activity of these constructs was evaluated using an in vitro rechallenge luciferase assay in ROR1 expressing JeKo-1 cells and ROR1(negative) controls. Results: The 2G 41BB-z construct with V H-V L scFv orientation and a long linker (V H-L-V L) showed optimal cytotoxicity with a CAR expression level in T cells of 36% (Range 28-49% for other constructs, Figure 1B-C). The V H-L-V L 41BB-z construct was evaluated comparatively using a rechallenge cytotoxic assay with 3G constructs that expressed CD28, ICOS, or OX40 signaling domains using JeKo-1 and ROR1(negative) target cells as controls. All the tested constructs showed specific ROR1 medicated cytotoxicity. CD28-41BB-z and ICOS-41BB-z showed the lowest cytotoxicity levels during the Day 1 of the repetitive rechallenge. However, the cytotoxicity levels of those constructs gradually increased during the 7 days of rechallenge and were closed to the levels induced by the 2G- 41BB-z construct (>80% of cytotoxicity). There were no significant differences in CAR T cells subsets generated by the different constructs during the 7 days of rechallenge with a predominance of effector memory phenotype (CCR7-, CD45RA-) and no difference in PD1 expression. Conclusions: Our results demonstrate that optimization of the CAR constructs enhances T-cell effector function and cytotoxicity against ROR1+ target cells. In previous studies, 3G CARs have shown longer persistence of the transduced T cells in peripheral blood, sustained and regulated cellular activation, improved solid tumor infiltration, and positive modulation of the tumor microenvironment. Our preclinical in vitro optimization demonstrates strategies to generate 3G constructs with a progressive and modulated cytotoxic profile that may confer benefits when tested in vivo in terms of enhanced persistence and lower adverse events profile. Additional experiments in vivo will be presented during the meeting to corroborate our findings. Figure 1 Figure 1. Disclosures No relevant conflicts of interest to declare.
- Published
- 2021
6. Management of Patients with Aggressive B Cell Non-Hodgkin Lymphoma after Relapse from Axicabtagene Ciloleucel: Single Center Real-World Experience
- Author
-
Mohamed A. Kharfan-Dabaja, Allison C. Rosenthal, Paula A Lengerke Diaz, Megan Melody, Eider F. Moreno Cortes, Januario E. Castro, and Jose V. Forero
- Subjects
Bendamustine ,medicine.medical_specialty ,business.industry ,Immunology ,Salvage therapy ,Cell Biology ,Hematology ,medicine.disease ,Single Center ,Biochemistry ,Gastroenterology ,chemistry.chemical_compound ,Cytokine release syndrome ,chemistry ,Obinutuzumab ,Internal medicine ,medicine ,Rituximab ,business ,Survival analysis ,medicine.drug ,Lenalidomide - Abstract
Background: Chimeric Antigen Receptor (CAR) T-cell therapy has changed the treatment landscape for patients with Non-Hodgkin Lymphoma (NHL). Despite the excellent responses in relapsed or refractory (R/R) aggressive NHL (aNHL), the outcome of patients (pts) that fail CAR T-cell therapy remains poor, and there is not a clear path for management of their disease. Methods: We conducted a retrospective analysis of aNHL pts treated with axicabtagene ciloleucel (axi-cel) at the Mayo Clinic campuses in Arizona and Florida between June 2018 and August 2020. We evaluated the predisposing factors, management, toxicities, and response after CAR T-cell therapy failure. Statistical calculations using parametric tests were performed, and survival curves were estimated using the Kaplan-Meier method and compared statistically using the log-rank test and Pearson's correlation. Results: Thirty-four pts with aNHL received axi-cel. The median age was 53 years [IQR 42-63], and 62% were male. All pts received inpatient axi-cel infusions and the median length of hospital stay was 14 days (IQR: 11-17). Cytokine Release Syndrome (CRS) was observed in 91% of pts (3% grade ≥3), while Immune Effector Cell Associated Neurotoxicity Syndrome (ICANS) was observed in 41% (24% grade ≥3). At day 30 response assessment, 16 pts (47%) had complete responses (CR), 9 (26%) had a partial response (PR), 4 (12%) had stable disease (SD), 4 (12%) showed progression with primary refractory disease (PD) and 1 (3%) died before assessment due to grade 5 ICANS (Table 1). After a median follow-up of 178 days, we observed PD in 12 (35%) pts. The median time-to-progression was 72 days (IQR 58-93) and most of the pts (83%) progressed during the first 3 months. None of the patients with more than 5 months of sustained response developed progression of disease. The likelihood of progression during the first 6 months after axi-cel infusion was 19%, 57%, 50% for pts that initially achieved a CR, PR, SD, respectively. Expression of CD19 was observed in 66% (4/6) of pts with available biopsies after axi-cel suggesting a failure mechanism other than antigen escape. The mortality rate of the R/R aNHL group was 58% with a median survival time of 83 days (IQR: 50-109). There was no association between age, stage, number of previous therapies, time from previous therapy to axi-cel infusion, time from apheresis to infusion, use of tocilizumab, or steroids with progression of disease. Of note, no correlation between CRS or ICANS with progression of disease was found (2-way ANOVA test F (1, 4) = 3.802, p=0.1230). Maintaining a response to axi-cel treatment (CR, PR, or SD) for ≥ 3 months was a strong predictor of durable response with an HR of 0.05 (p= Eleven R/R pts received subsequent therapies with a median time to retreatment of 76 days. Those treatments included: Radiotherapy (n=7), pembrolizumab (n=3), polatuzumab-rituximab with (n=3) and without (n=1) bendamustine, obinutuzumab with (n=1) or without (n=1) lenalidomide, Hyper-CVAD (n=2), R-GemOx (n=1), rituximab with lenalidomide (n=1) and intrathecal methotrexate (n=1). Only 2 (17%) patients have responded to salvage therapy achieving PR (one patient treated with radiotherapy and the other with rituximab-lenalidomide after two other salvage therapies). Conclusion: Our experience demonstrates the majority of aNHL patients respond to axi-cel. If patients maintain their response for more than 3 months, the likelihood of progression is very low - 15%. Similar to what has been previously reported in the literature, our series showed that 35% of patients progressed after axi-cel, and subsequently have a poor prognosis with median survival after a relapse of only 83 days (IQR: 50-109). Therapy options following axi-cel were limited due to severe cytopenias, only 2 of 11 patients have responded to salvage therapy, suggesting that conventional treatments are probably not effective/safe in this high-risk group of patients. Interestingly, the majority of R/R pts with available biopsies showed persistent CD19 expression suggesting that CAR T-cell exhaustion, poor in vivo expansion, and inhibitory signals of the tumor microenvironment may contribute to resistance. Additional strategies for monitoring of axi-cel persistence and its immunophenotypic profile could be helpful for prognosis and management of CAR T-cell pts receiving axi-cel. Disclosures Kharfan-Dabaja: Daiichi Sankyo: Consultancy; Pharmacyclics: Consultancy. Castro:Fate Therapeutics: Research Funding; Kite Pharma: Research Funding; Pharmacyclics: Research Funding.
- Published
- 2020
7. Long-Term Sustained Responses Following Ibrutinib Discontinuation after Frontline Therapy with Obinutuzumab Plus Ibrutinib in Patients with Chronic Lymphocytic Leukemia
- Author
-
Carlos I. Amaya-Chanaga, Juliana Velez-Lujan, Januario E. Castro, Paula A Lengerke Diaz, Michael Y. Choi, Eider F. Moreno Cortes, Thomas J. Kipps, and Jose V. Forero
- Subjects
Oncology ,medicine.medical_specialty ,business.industry ,Chronic lymphocytic leukemia ,Immunology ,Cell Biology ,Hematology ,medicine.disease ,Biochemistry ,Discontinuation ,chemistry.chemical_compound ,chemistry ,Obinutuzumab ,Ibrutinib ,Internal medicine ,Medicine ,In patient ,business - Abstract
Single oral targeted therapies have emerged as a standard of care in chronic lymphocytic leukemia (CLL). However, accessibility, side effects, and financial burden associated with long term administration limit their clinical use. Mainly, it is unclear in what clinical situation discontinuation of oral therapy can be recommended. The combination of type II anti-CD20 antibody obinutuzumab-Gazyva® with ibrutinib (GI) has shown a significant progression-free survival benefit in patients (pts) with CLL, including those with high-risk genomic aberrations. We conducted a phase 1b/2, single-arm, open-label trial to evaluate the safety and efficacy of GI as first-line treatment in 32 CLL pts. We report the outcome in pts that discontinued ibrutinib (either after 3 years of sustained complete response (CR) as stipulated in the clinical protocol, or due to other reasons). CLL pts enrolled in this protocol were ≥65 years old, or unfit/unwilling to receive chemotherapy. Pts received GI for six cycles, followed by daily single-agent ibrutinib. The protocol was designed to ensure that pts with a sustained CR after 36 months were allowed to discontinue ibrutinib. The median age was 66 years (IQR 59-73), and 6% of the evaluated pts had 17p deletion. All pts were able to complete the six planned cycles of obinutuzumab. The combination regimen was well-tolerated, and the most common adverse events (>5% CTCAE grade 3-4) were neutropenia, thrombocytopenia, and hyperglycemia. The rate and severity of infusion-related reactions (IRR) were much lower than expected (Grade≥ 3, 3%), and pts without IRR had lower serum levels of cytokines/chemokines CCL3 (P=0.0460), IFN-γ (P=0.0457), and TNF-α (P=0.0032) after infusion. The overall response rate was 100%, with nine pts (28%) achieving a CR, and four pts (12.5%) with undetectable minimal residual disease (uMRD) in the bone marrow, defined as Sixteen pts have completed a long-term follow-up of 36 months. Six pts showed CR, with three of them achieving uMRD in the bone marrow. Ten of these pts were in PR, and only one had disease progression and started treatment for symptomatic stage I disease with obinutuzumab plus venetoclax. In total, thirteen pts (41%) have stopped ibrutinib, with a median time on treatment prior to discontinuation of 35 months. Five (16%) of these pts had CRs and discontinued after 36 months. Eight additional pts (25%) had PRs and discontinued ibrutinib without being eligible: three pts discontinued prior to 36 months due to toxicities, and five pts discontinued after 36 months (3 due to side effects, and 2 due to financially driven decision). One patient eligible to discontinue ibrutinib, decided to remain on treatment despite sustained CR. After a median follow up time following ibrutinib discontinuation of 8 months (IQR 3.5-17), only two out of 13 pts have progressed (10 and 17 months after Ibrutinib discontinuation). None of the pts that stopped ibrutinib after achieving a CR have shown signs of disease progression. Of note, the pharmaceutical sponsor provided ibrutinib for the first 36 months, after which pts or their insurer became financially responsible. This particular scenario could bias the discontinuation pattern compared to a real world experience. It also provided us with a perspective about diverse factors affecting the treatment choices of pts. In summary, the obinutuzumab plus ibrutinib combination therapy was well-tolerated, with a much lower IRR rate. Efficacy compares favorably with historical controls with all pts responding to therapy, no deaths associated with treatment or disease progression, and a longer than expected time-to-progression after discontinuation of ibrutinib. The rate of ibrutinib discontinuation was higher than reported in the literature, most likely influenced by the protocol design and financial decisions driven by the switch from sponsor-provided ibrutinib to insurance or self-paid medication. Our observations regarding safety, efficacy and lack of disease progression after ibrutinib discontinuation are encouraging, and warrant confirmation in long-term prospective studies. Clinicaltrials.gov Identifier NCT02315768. Funding: Pharmacyclics LLC. Disclosures Choi: AbbVie: Consultancy, Speakers Bureau. Amaya-Chanaga:AbbVie: Ended employment in the past 24 months, Other: Research performed while employed as an investigator of this study at UCSD.. Kipps:Pharmacyclics: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding. Castro:Kite Pharma: Research Funding; Pharmacyclics: Research Funding; Fate Therapeutics: Research Funding.
- Published
- 2020
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.