Abhijeet Kumar
- Associate Professor, Medicine - (Clinical Scholar Track)
Contact
- (520) 626-6453
- Leon Levy Cancer Center, Rm. 2301
- Tucson, AZ 85724
- akumar1@arizona.edu
Degrees
- M.B.B.S.
- Sikkim Manipal Institute of Medical Sciences, Gangtok, India
Work Experience
- The John Hopkins Hospital (2012)
- St. Agnes Hospital (2011 - 2014)
- Community (2007 - 2011)
Awards
- The Krishnadasan Award for Educational Excellence
- UACC, Spring 2022
- ECFMG IAN Advisor
- Summer 2016
- Bressler-Alpert travel award
- Spring 2016
- Chief Resident (3rd Year)
- UofA, Spring 2013
- Intern of the Year
- COM, Summer 2011
Licensure & Certification
- NPI #1134409923 (2011)
- USMLE Step 3 - 224/99 (2010)
- Arizona Medical License, AMB (2017)
- American Board of Internal Medicine, ABIM (2014)
- ECFMG Certificate # 0-872-401-5 (2010)
- USMLE Step 1 - 241/99 (2010)
- USMLE Step 2 - 247/99 (2010)
Interests
Research
Immunotherapy and resistance to immune checkpoint inhibition in lymphoid malignancies, modalities to overcome late relapses in patients with localized DLBCL, clinical trials using combination of aurora kinase inhibition and BTK or downstream signaling inhibition as an modality for relapsed refractory double hit double protein or ABC subtype DLBCL; Mechanisms Resistance to immune checkpoint inhibition is sarcomas.
Courses
No activities entered.
Scholarly Contributions
Chapters
- Korde, N., Kumar, A., Agarwal, A. B., & Sundararajan, S. (2016). MGUS, Smoldering Myeloma and Plasmacytoma. In Handbook of Hematologic Malignancies 1st Edition.
- Kumar, A., Krishnadasan, R., & Sundararajan, S. (2016). T-cell Prolymphocytic Lymphoma. In Handbook of Hematologic Malignancies 1st Edition.
Journals/Publications
- Li, D., Shields, A. F., Mamdani, H., Travis, P., Wright, G., Akerley, W., Spira, A., Daniels, G., Merchan, J., Kumar, A., Lim, E., Egelston, C., Flores, O., Hamilton, S., Austria, J., Seiz, A., Yavrom, S., Fein, S., Byon, J., , Woodard, P., et al. (2024). Abstract CT182: Oncolytic virus CF33-hNIS for the treatment of advanced cancer. Cancer Research, 84(7_Supplement), CT182-CT182. doi:10.1158/1538-7445.am2024-ct182
- Sancho, J. M., Abrisqueta, P., Kumar, A., Cordoba, R., Tani, M., Langmuir, P., Rappold, E., Liu, T., & Lopez-Guillermo, A. (2024). Safety and efficacy of parsaclisib in combination with rituximab, bendamustine + rituximab, or ibrutinib in patients with previously treated B-cell lymphoma: analysis of a phase 1 dose-finding study (CITADEL‑112). Leukemia & lymphoma, 65(7), 911-921.More infoParsaclisib, a potent and highly selective phosphoinositide 3-kinase δ inhibitor, has shown clinical activity in relapsed/refractory (R/R) B-cell lymphoma. The phase 1 CITADEL-112 (NCT03424122) study assessed safety and efficacy of parsaclisib in combination with investigator choice standard of care (SOC; rituximab [Treatment A], rituximab plus bendamustine [Treatment B], or ibrutinib [Treatment C]) in 50 patients with R/R B-cell lymphoma. The most common treatment-emergent adverse events included neutropenia (62.5%, 50.0%, and 50.0% of patients in Treatments A, B, and C, respectively); diarrhea (37.5%) and anemia (31.3%) in Treatment A; abdominal pain, asthenia, diarrhea, and nausea (each 33.3%) in Treatment B; and increased alanine and aspartate aminotransferase (each 37.5%) in Treatment C. Objective responses were observed in 13 patients (81.3%) in Treatment A, 10 (55.6%) in Treatment B, and 8 (50.0%) in Treatment C. Parsaclisib combined with SOC therapies had an expected safety profile and promising efficacy in patients with R/R B-cell lymphomas.
- Sancho, J., Abrisqueta, P., Kumar, A., Cordoba, R., Tani, M., Langmuir, P., Rappold, E., Liu, T., & Lopez-Guillermo, A. (2024). Safety and efficacy of parsaclisib in combination with rituximab, bendamustine + rituximab, or ibrutinib in patients with previously treated B-cell lymphoma: analysis of a phase 1 dose-finding study (CITADEL‑112). Leukemia & Lymphoma, 1-11. doi:10.1080/10428194.2024.2331626
- Soumerai, J. D., Diefenbach, C. S., Jagadeesh, D., Asch, A., Kumar, A., Tsai, M. L., Jandl, T. A., Lossos, I. S., Kenkre, V. P., Awan, F., Novotny, W., Huang, J., Miao, L., Rajagopalan, P., Ghalie, R. G., & Zelenetz, A. D. (2024). Safety and efficacy of zandelisib plus zanubrutinib in previously treated follicular and mantle cell lymphomas. British Journal of Haematology. doi:10.1111/bjh.19419
- Soumerai, J. D., Diefenbach, C. S., Jagadeesh, D., Asch, A., Kumar, A., Tsai, M. L., Jandl, T. A., Lossos, I. S., Kenkre, V. P., Awan, F., Novotny, W., Huang, J., Miao, L., Rajagopalan, P., Ghalie, R. G., & Zelenetz, A. D. (2024). Safety and efficacy of zandelisib plus zanubrutinib in previously treated follicular and mantle cell lymphomas. British journal of haematology, 204(5), 1762-1770.More infoThe combination of the phosphatidylinositol 3-kinase delta (PI3Kδ) inhibitor zandelisib with the Bruton's tyrosine kinase (BTK) inhibitor zanubrutinib was hypothesized to be synergistic and prevent resistance to single-agent therapy. This phase 1 study (NCT02914938) included a dose-finding stage in patients with relapsed/refractory (R/R) B-cell malignancies (n = 20) and disease-specific expansion cohorts in follicular lymphoma (FL; n = 31) or mantle cell lymphoma (MCL; n = 19). The recommended phase 2 dose was zandelisib 60 mg on Days 1-7 plus zanubrutinib 80 mg twice daily continuously in 28-day cycle. In the total population, the most common adverse events (AEs; all grades/grade 3-4) were neutropenia (35%/24%), diarrhoea (33%/2%), thrombocytopenia (32%/8%), anaemia (27%/8%), increased creatinine (25%/0%), contusion (21%/0%), fatigue (21%/2%), nausea (21%/2%) and increased aspartate aminotransferase (24%/6%). Three patients discontinued due to AEs. The overall response rate was 87% (complete response [CR] = 33%) for FL and 74% (CR = 47%) for MCL. The median duration of response and progression-free survival (PFS) were not reached in either group. The estimated 1-year PFS was 72.3% (95% confidence interval [CI], 51.9-85.1) for FL and 56.3% (95% CI, 28.9-76.7) for MCL (median follow-up: 16.5 and 10.9 months respectively). Zandelisib plus zanubrutinib was associated with high response rates and no increased toxicity compared to either agent alone.
- Menghani, S. V., Diaz-Hanson, J. P., Heimbigner, A., Wakefield, C., Fuchs, D., Reveles, C. Y., Spier, C., Amaraneni, A., & Kumar, A. (2023). Peripheral T-Cell Lymphoma in a Patient Previously Diagnosed With Sarcoidosis. Journal of hematology, 12(6), 272-276.More infoSarcoidosis is a multisystem disorder characterized by granulomatous inflammation on histopathological evaluation. Diagnosis of sarcoidosis requires thorough elimination of malignancy and alternative causes of noncaseating granulomatous inflammation. Sarcoidosis and several subtypes of lymphoma have similar clinical presentations and can potentially have similar histopathological findings. Patients with a histopathology-confirmed diagnosis of sarcoidosis are at higher risk of developing malignancies. In this report, we present a case of a 64-year-old male diagnosed with sarcoidosis 2 years before presenting to the emergency department with a 4-month history of generalized weakness, cough, and very high fever. After a thorough workup involving cervical lymph node biopsy and bone marrow biopsy, he was diagnosed with peripheral T-cell lymphoma, not otherwise specified (PTCL, NOS). Due to the patient's current lymphoma diagnosis and features noted on pathology, a retrospective review of the prior biopsy specimen was performed, finding similar hematopathological features on both initial lymph node biopsy diagnosing sarcoidosis and current biopsies diagnosing lymphoma. Given these findings, our patient likely had early manifestation of PTCL misdiagnosed as sarcoidosis. In summary, lymphoma should be considered in all patients with suspected sarcoidosis, especially those who do not respond to treatment or who present with persistent hematological abnormalities.
- Seymour, J. F., Cheah, C. Y., Parrondo, R., Thompson, M. C., Stevens, D. A., Lasica, M., Wang, M. L., Kumar, A., Trotman, J., Alwan, M., Ding, W., By, K., Tariq, B., Chen, X., Fabre, S., Paik, J., Agarwal, A., & Tam, C. S. (2023). First Results from a Phase 1, First-in-Human Study of the Bruton's Tyrosine Kinase (BTK) Degrader Bgb-16673 in Patients (Pts) with Relapsed or Refractory (R/R) B-Cell Malignancies (BGB-16673-101). Blood, 142(Supplement 1), 4401-4401. doi:10.1182/blood-2023-180109
- Abrisqueta, P., Cordoba, R., Kumar, A., Langmuir, P., Liu, T., Lopez-Guillermo, A., Rappold, E., Sancho, J., & Tani, M. (2022). Safety and Efficacy of Parsaclisib in Combination with Rituximab, Bendamustine + Rituximab, or Ibrutinib in Patients with Previously Treated B-Cell Lymphoma: Analysis of a Phase 1 Dose-Finding Study (CITADEL-112). Blood, 140(Supplement 1), 9328-9330. doi:10.1182/blood-2022-159703More infoBackground: Rituximab (RIT)-based chemoimmunotherapy regimens are backbone treatment for indolent (follicular, marginal zone) and aggressive (diffuse large B-cell, mantle cell) B-cell lymphomas. For relapsed or refractory (R/R) disease, anti-CD20 antibodies can be combined with chemotherapy or targeted therapies, such as inhibitors of Bruton's tyrosine kinase (eg, ibrutinib) and phosphoinositide 3-kinase (PI3K). Parsaclisib is a potent and highly selective next-generation PI3Kδ inhibitor. In a preliminary safety analysis of CITADEL-112 (NCT03424122), an open-label phase 1 study of parsaclisib plus investigator choice standard of care (RIT, RIT + bendamustine [BEN], or ibrutinib [IBR]) in patients (pts) with R/R B-cell lymphoma, manageable tolerability was observed (Sancho JM, et al. HemaSphere 2022;6 Suppl 3:P1102). Here we present updated safety and initial efficacy data from CITADEL-112. Methods: Enrolled pts were ≥18 years and had histologically confirmed DLBCL, FL, MCL, or MZL, ECOG performance status of 0-2, were R/R to ≥1 (≥2 for FL) prior systemic therapy, and ineligible for stem cell transplantation. Pts received parsaclisib 20 mg orally once daily (QD) for 8 weeks then 20 mg once weekly (QW) in combination with either RIT 375 mg/m2 IV QW for 4 doses in cycle 1 (± an additional 4 QW doses) in Cohort A, or RIT 375 mg/m2 IV on day 1 + BEN 90 mg/m2 on day 1 and day 2 of each 28-day cycle for ≤6 cycles in Cohort B, or IBR 560 mg QD in Cohort C. A 3+3 design with dose de-escalation identified the maximum tolerated dose of QD parsaclisib within each treatment regimen independently. Pts received treatment until disease progression, unacceptable toxicity, or withdrawal. The primary endpoint was safety and tolerability. Investigator-determined objective response rate (ORR) and duration of response (DOR) were exploratory endpoints. Results: At data cutoff (January 14, 2022), 50 pts were treated (16 pts each in Cohorts A and C, 18 pts in Cohort B) and 5 pts were ongoing treatment (all in Cohort B). Most pts had received ≥2 prior systemic treatments; the most common disease subtypes were FL in Cohort A and DLBCL in Cohorts B and C (Table 1). Median (range) dose of QD parsaclisib was 20.0 (13.3-20.0), 17.9 (2.8-20.0), and 20.0 (7.1-20.0) mg/day in Cohorts A, B, and C, respectively, and median (range) overall duration (QD+QW) of parsaclisib treatment was 5.1 (0.5-22.9), 9.3 (0.5-33.7), and 9.3 (0.6-37.7) months. The most common reasons for discontinuation were progressive disease (75.0%, 44.4%, and 56.3% in Cohorts A, B, and C, respectively) and adverse events (AEs; 12.5%, 16.7%, and 6.3%, respectively). One pt in Cohort B experienced a dose-limiting toxicity of grade 4 neutropenia for 14 days. All pts experienced ≥1 treatment-emergent AE (TEAE); most common any-grade and grade ≥3 TEAEs are shown in Table 2. Grade ≥3 and serious TEAEs were experienced by 75.0% and 37.5% of pts, respectively, in Cohort A, 88.9% and 33.3% of pts in Cohort B, and 62.5% and 50.0% of pts in Cohort C. Serious TEAEs occurring in >1 pt were COVID-19, diarrhea, and pneumonia (n=2 each) in Cohort A, and atrial fibrillation (n=2) in Cohort C. TEAEs with fatal outcome occurred in 2 pts in Cohort A (COVID-19 and COVID-19 pneumonia [n=1], interstitial lung disease [n=1]) and 1 pt in Cohort C (COVID-19 pneumonia, septic shock, acute kidney injury, respiratory failure). TEAEs leading to discontinuation of parsaclisib occurred in 2 pts in Cohort A (diarrhea, streptococcal pneumonia [n=1 each]), 3 pts in Cohort B (neutropenia, maculopapular rash [n=1 each], diarrhea and renal failure [n=1]), and 1 pt in Cohort C (thrombocytopenia). TEAEs leading to parsaclisib dose interruption or reduction occurred in 75.0% and 18.8% of pts, respectively, in Cohort A, 66.7% and 27.8% of pts in Cohort B, and 56.3% and 18.8% of pts in Cohort C. ORRs (95% confidence interval [CI]) were 81.3% (54.4-96.0), 55.6% (30.8-78.5), and 50.0% (24.7-75.3) for Cohorts A, B, and C, respectively; 18.8%, 33.3%, and 12.5% of pts had a complete metabolic response/complete response, respectively (Table 1). Median (95% CI) DOR was 5.2 months (1.7-15.8) for Cohort A, not reached (12.1-not evaluable [NE]) for Cohort B, and 13.4 months (0.7-NE) for Cohort C. Conclusion: Parsaclisib 20 mg QD for 8 weeks followed by 20 mg QW in combination with RIT, RIT+BEN, or IBR in pts with R/R B-cell lymphomas had a manageable tolerability profile with no unexpected safety concerns, and demonstrated promising efficacy. Figure 1View largeDownload PPTFigure 1View largeDownload PPT Close modal
- Amaraneni, A., Dashkevych, U., Gowin, K. L., Husnain, M., Katsanis, E., Khurana, S., Krishnadasan, R., Kumar, A., Munugala, N., & Pu, J. J. (2022). Role of Anakinra in the Management of Steroid Refractory High Grade Immune Effector Cell-Associated Neurotoxicity Syndrome (ICANS) after Anti-CD 19 CAR-T Cell Therapy, a Single Center Experience. Blood, 140(Supplement 1), 12753-12754. doi:10.1182/blood-2022-168240More infoBackground: Chimeric antigen receptor (CAR) T-cell immunotherapy is a paradigm shifting treatment option for multiple refractory hematologic malignancies. Immune effector cell-associated neurotoxicity syndrome (ICANS) is a serious adverse effect (AE) associated with CAR T-cell therapy. To date, corticosteroids remain the cornerstone treatment of ICANS. Recently, limited clinical data have suggested that the addition of anakinra may be beneficial in the treatment of corticosteroid refractory severe ICANS. Methods: We performed a retrospective chart review of patients with a diagnosis of refractory B cell lymphoma treated with CART-cell therapy at the University of Arizona Cancer Center (UACC), who subsequently developed severe ICANS (grade 3-4). We report five cases in which anakinra (at the dose of 100 mg daily) was used as an adjunct therapy to high dose steroid treatment in managing severe ICANS. Results: A total of 33 patients received CART-cell therapy at UACC from 01/2019-07/2022. The incidence of severe ICANS was 21% (7/33). Baseline characteristics of 7 patients who developed high grade ICANS are summarized in Table 1. The median age was 65 (range 51-70). 5 patients were treated with Tisagenlecleucel, 1 with Axicabtagene ciloleucel and 1 with Brexucabtagene autoleucel CAR-T cell therapy. The median time to onset of severe ICANS was 6 days (range 1-13 days). Two out of seven patients (29%) developed grade 3 ICANS which resolved quickly with corticosteroids in 1-3 days. Five out of seven patients (71%) developed grade 4 ICANS, requiring ICU admission and high dose corticosteroids. Patients received at least 3 days of high dose corticosteroids with methylprednisolone 500 mg IV twice a day before anakinra was added for steroid refractory ICANS. On average, patients received ~ 9 doses of corticosteroids prior to anakinra administration. After addition of anakinra, three patients had a complete resolution of ICANS, two patients had partial improvement in their ICANS (grade 1-2). For the three patients with complete resolution of ICANS, the median number of anakinra doses were 4, median duration of severe ICANS (post-anakinra) was 3 days and the median time in ICU was 3 days. Clinical course was complicated for the other two patients with acute hypoxic respiratory failure in one patient and E. coli bacteremia, fulminant pseudomembranous colitis s/p colectomy, and septic shock in the second patient leading to death on days +23 and +40 respectively. Conclusion: Although limited in number, our analysis suggests anakinra may be effective and safe in steroid refractory ICANS. ICANS resolved completely in three out of five patients following administration of anakinra who were refractory to high dose corticosteroids, and partially improved to grade 1-2 in two patients. No significant side effects attributed to anakinra were observed. Future randomized controlled studies are needed to better understand the overall efficacy and the ideal timeline for administration. Figure 1View largeDownload PPTFigure 1View largeDownload PPT Close modal
- Chineke, I., Hassab, M., Husnain, M., & Kumar, A. (2022). Anti-CD 19 CAR-T Cell Therapy with Tisagenlecleucel for Ritcher Syndrome. Blood, 140(Supplement 1), 12796-12797. doi:10.1182/blood-2022-167450More infoAbout 2-10 percent of patients with chronic lymphocytic leukemia (CLL) will transform into an aggressive large cell lymphoma, called Ritcher syndrome (RS). The transformation is to DLBCL in 90-95% of cases and Hodgkin lymphoma in the remaining 5-10%. Current treatment strategies that typically mirror DLBCL have not significantly improved the historically dismal outcomes of RS. Chimeric antigen receptor-modified T (CAR-T) cells have revolutionized the treatment of B-cell malignancies and are currently approved for relapsed/refractory DLBCL. The impact of CAR-T cell therapy in the management of RS is limited by the small number of affected patients and published reports. We report the outcomes in 2 patients with RS treated with tisagenlecleucel (Tisa-cel) at our institution. Patient 1. This is a 75-year-old lady diagnosed with CLL at the age of 64, Rai stage 1, trisomy 12, mutated IGVH. She was on observation until she became symptomatic 5 yrs. after diagnosis and had a biopsy which showed CLL with RS, fluorescent in-situ hybridization (FISH) negative for C-MYC, BCL2 and BCL6. She was treated with RCHOP x 6 cycles with complete remission (CR) on PET. She received high dose chemotherapy with R-BEAM followed by autologous stem cell transplant, followed by 4 cycles of maintenance brentuximab given every 3 weeks for her CD30 positive large B cell lymphoma. About 5 yrs. later, surveillance PET showed hypermetabolic cervical lymph nodes and excisional biopsies showed CLL and DLBCL, non-germinal center type, concerning for relapsed RS. She was referred to us for CAR-T cell therapy evaluation. Bone marrow biopsy was negative for marrow involvement, minimal residual disease (MRD) by ClonoSEO was positive. She received lymphodepleting chemotherapy with fludarabine and cyclophosphamide followed by CAR-T cell infusion with tisa-cel 5x10^8 anti CD19 CAR-T cells on 4/18/22. Treatment was complicated by grade 1-2 cytokine release syndrome (CRS) (treated with tocilizumab and dexamethasone) requiring brief hospitalization. Day 90 PET showed CMR and bone marrow biopsy was negative for disease, MRD negative. She continues to be in CR 100 days post CAR-T cell therapy. Patient 2. A 63 years-old lady originally diagnosed with CLL in 2016 at the age of 57, with 17p deletion, 13q deletion and IgHV unmutated. She was started on ibrutinib shortly after due to a short lymphocyte doubling time. Three years later (in 2020), she developed worsening lymphadenopathy while on ibrutinib, underwent excisional biopsy and this showed RS with TP53 mutation, 17p deletion, gain of 13q, BCL2 positive. Venetoclax was added to ibrutinib and later, 2 cycles of Obinutuzumab were also given. Subsequent PET 7 months after showed disease progression with a lugano score of 5. She received 6 cycles of RCHOP in addition to venetoclax/ibrutinib but end of treatment PET showed disease progression. She was then referred to us for CAR-T cell evaluation. She received lymphodepleting chemotherapy with fludarabine and cyclophosphamide followed by CAR-T cell infusion with tis-cel 3.7 x 10^8 cells anti CD19 CAR-T cells on 6/7/21. This was complicated by grade 2 CRS that was treated with tocilizumab. Day 30 PET showed PR. Ibrutinib was resumed and the patient received radiation to right cervical lymph nodes. Day 100 PET reaffirmed PR and she subsequently underwent FluCyTBI RIC followed by haploidentical alloSCT from her daughter on 09/29/21. On day 180 post-transplant, the patient remained in CR by PET and bone marrow biopsy. She continues to be in CR now 1 year since her CAR-T cell infusion. Conclusion CAR-T cell therapy with Tisagenlecleucel shows promising activity in patients with RS. It is an evolving treatment strategy and more follow up is needed to assess the durability of the responses observed in these patients.
- Dhaliwal, A., Husnain, M., Kumar, A., Mann, S., & Persky, D. O. (2022). Tisagenlecleucel for secondary CNS lymphoma, a case series.. Journal of Clinical Oncology, 40(16_suppl), e19507-e19507. doi:10.1200/jco.2022.40.16_suppl.e19507More infoe19507 Background: Secondary CNS lymphoma (SCNSL) in setting of relapsed, refractory (R/R) B cell lymphoma has unmet needs as this disease state has a poor prognosis with a reported median survival of 2 to 5 months. Historically, the mainstay of treatment for SCNSL in setting of R/R B cell lymphoma included high dose methotrexate, whole brain radiation (WBRT), and/or chemotherapy followed by autologous stem cell transplant. More recently, chimeric antigen receptor (CAR)-T cell therapy has gained attention as a new modality of therapy for patients with R/R SCNSL. The first two CAR-T cell products to gain FDA approval for R/R B cell lymphoma were Axicabtagene ciloleucel and Tisagenlecleucel. In the initial studies for these products, patients with SCNSL were excluded due to concerns for immune effector cell-associated neurotoxicity syndrome (ICANS) and cytokine release syndrome (CRS). Retrospective data has shown that patients with R/R SCNSL treated with Axicabtagene ciloleucel or Tisagenlecleucel could achieve high response rates; however, the data on the durability of response remains unclear. Recently, Lisocabtagene maraleucel, another CAR-T cell product, was approved for the treatment of R/R B cell lymphoma. The TRANSCEND trial which led to Lisocabtagene maraleucels approval did not exclude patients with SCNSL; however, only 3% of the patients had SCNSL. As such, literature on the durability of response of CAR-T cell therapy in R/R SCNSL still remains sparse. Here, we share our single institution experience of patients with R/R SCNSL treated with Tisagenlecleucel, an anti- CD19CAR-T cell therapy. Methods: For this observational study, patients with R/R SCNSL were identified and placed on a follow up list. Data was collected from May 2021 till December 2021. Data was analyzed for incidence of ICANS, CRS, and for duration of response. Results: A total of 3 patients with SCNSL were identified and treated at our center with Tisagelecleucel. One patient achieved complete response 30 days post CAR-T therapy but the day +100 imaging revealed progression of disease and was placed on subsequent line of therapy. One patient had partial response with progression of disease noted on day +60 imaging and died from complications of disease progression. One patient died from complications of CNS toxicity after receiving CAR-T therapy. Two patients had ICANS grade 4 and CRS grade 1 toxicities. One patient had no CNS or CRS toxicity. Conclusions: Our series is limited by the number of patients; however, suggests that though CAR-T therapy may result in disease response in R/R SCNSL the duration of response may be limited and that such patients may require other therapies such as autologous stem cell transplant to improve long term prognosis.
- Dhaliwal, A., Zandu, M. K., Khurana, S., Kumar, A., Pu, J. J., Katsanis, E., & Husnain, M. (2022). Anti CD19 CAR-T Cell Therapy Can Clear Minimal Residual Disease in Bone Marrow of Patients with Relapsed or Refractory B Cell Non-Hodgkin's Lymphomas. Blood, 140(Supplement 1), 12760-12761. doi:10.1182/blood-2022-170620More infoBackground: Anti CD19 Chimeric Antigen Receptor T (CAR-T) cell therapy is an effective treatment option for patients with relapsed/refractory B-cell lymphomas. There is limited data on the impact of CAR-T cell therapy on measurable residual disease (MRD) in the bone marrow of these patients. We hereby report CAR-T cell therapy's efficacy to treat bone marrow MRD in patient with relapsed/refractory B-cell lymphomas. Methods: In this retrospective review, patients receiving CAR-T cell therapy for relapsed/refractory B-cell lymphomas who had bone marrow MRD evaluation pre-and post-CAR-T infusion at the University of Arizona Cancer Center (UACC) were analyzed. MRD testing was performed via ClonoSEQ (Adaptive Biotechnologies) on bone marrow aspirates prior to CAR-T cell infusion. Patients with MRD+ (10^6) disease had serial MRD evaluations at day+30, +90, +180, +365 post CAR-T cell infusion. Results: Thirty-three patients with B-cell lymphomas received CAR-T cell therapy at UACC from January 2019 to July 2022. Bone marrow MRD pre-and post-CART was evaluated in ten patients. Baseline characteristics of these patients are outlined in Table 1. Seven (70%) were female, five (50%) had diffuse large B cell lymphoma (DLBCL), two (20%) patients had Richter's syndrome, two patients had transformed DLBCL from follicular lymphoma (FL), one patient had FL grade 3A. Nine (90%) patients were treated with Tisagenlecleucel and one (10%) patient received Axicabtagene ciloleucel CAR-T product. Of the ten patients who had MRD testing done pre-and post-CAR-T, seven (70%) had MRD positive disease prior to treatment. Five of the seven patients converted to MRD negative 10^6 post-CAR-T cell infusion. One patient tested MRD positive below the level of detection on day +30 bone marrow aspirate. One patient is still MRD positive 6 months post-CAR-T infusion but the depth of response continues to improve from MRD positive at 10^4 to MRD negative at 10^5, 6 months post-CAR-T infusion. All MRD positive patients achieved complete metabolic response (CMR) based on the positron emission tomography scans. Conclusion: CAR-T cell therapy has the potential to treat minimal residual disease in patients with B-cell lymphoma. Depth of bone marrow response continues to improve over time. All patients continue to be in CMR. Long term follow-up will help determine if persistent MRD can be used as prognostic marker in B cell lymphomas post CAR-T cell therapy. Figure 1View largeDownload PPTFigure 1View largeDownload PPT Close modal
- O’Neil, B. H., Solis, W., Vidal-Cardenas, S., Uttamsingh, S., & Kumar, A. (2022). A First-in-Human Phase 1 Dose Escalation and Expansion Study of the Oral CARD11-BCL10-MALT1 Signaling Pathway Inhibitor XL114 in Patients with Non-Hodgkin's Lymphoma. Blood, 140(Supplement 1), 12055-12056. doi:10.1182/blood-2022-167824
- Sancho, J., Lopez-Guillermo, A., Abrisqueta, P., Kumar, A., Cordoba, R., Tani, M., Zhao, W., Rappold, E., Langmuir, P., & Mims, M. (2022). P1102: A PHASE 1 STUDY OF PARSACLISIB IN COMBINATION WITH RITUXIMAB, BENDAMUSTINE + RITUXIMAB, OR IBRUTINIB IN PATIENTS WITH PREVIOUSLY TREATED B-CELL LYMPHOMA (CITADEL-112): PRELIMINARY SAFETY RESULTS. HemaSphere, 6, 992-993. doi:10.1097/01.hs9.0000847276.06711.b7
- Soumerai, J. D., Diefenbach, C., Samaniego, F., Kumar, A., Tsai, M. L., Asch, A. S., Jagadeesh, D., Kenkre, V. P., Lossos, I. S., Salman, H., Awan, F. T., Liu, M., Ghalie, R., & Zelenetz, A. D. (2022). Safety and Efficacy of the PI3Kδ Inhibitor Zandelisib in Combination with the BTK Inhibitor Zanubrutinib in Patients with Relapsed/Refractory (R/R) Follicular Lymphoma (FL) or Mantle Cell Lymphoma (MCL). Blood, 140(Supplement 1), 189-191. doi:10.1182/blood-2022-157563
- Cardona, K., Chou, A. J., Copeland, T., D'Amato, G. Z., Demeure, M. J., Dizon, D. S., Elliott, A., Espejo Freire, A. P., Florou, V., Gibney, G. T., Groisberg, R., Khushman, M. M., Korn, W. M., Kumar, A., Lagos, G., Modiano, J., Riedel, R. F., Seeber, A., Trent, J. C., & von Mehren, M. (2021). Large scale multiomic analysis suggests mechanisms of resistance to immunotherapy in leiomyosarcoma.. Journal of Clinical Oncology, 39(15_suppl), 11512-11512. doi:10.1200/jco.2021.39.15_suppl.11512
- Copeland, T., Groisberg, R., Dizon, D. S., Elliott, A., Lagos, G. G., Seeber, A., Mehren, M. v., Cardona, K., Demeure, M. J., Riedel, R. F., Florou, V., Chou, A. J., Kumar, A., Modiano, J. F., Khushman, M., D’Amato, G. Z., Espejo-Freire, A. P., Korn, W. M., & Trent, J. C. (2021). Multiomic analysis to reveal distinct molecular profiles of uterine and nonuterine leiomyosarcoma.. Journal of Clinical Oncology, 39(15_suppl), 11555-11555. doi:10.1200/jco.2021.39.15_suppl.11555
- Davids, M. S., Roberts, A. W., Kenkre, V. P., Wierda, W. G., Kumar, A., Kipps, T. J., Boyer, M., Salem, A. H., Pesko, J. C., Arzt, J. A., Mantas, M., Kim, S. Y., & Seymour, J. F. (2021). Long-term Follow-up of Patients with Relapsed or Refractory Non-Hodgkin Lymphoma Treated with Venetoclax in a Phase I, First-in-Human Study. Clinical cancer research : an official journal of the American Association for Cancer Research, 27(17), 4690-4695.More infoWe previously reported a 44% overall response rate (ORR) with the oral BCL-2 inhibitor venetoclax in a phase I study of relapsed/refractory non-Hodgkin lymphoma (NHL). Complete response (CR) was observed in patients with mantle cell lymphoma [(MCL), 21%, = 6/28] and follicular lymphoma [(FL), 17%, = 5/29], and partial response (PR) noted in several patients with Waldenström macroglobulinemia (WM), and marginal zone lymphoma (MZL). Here, we report the long-term outcomes of these four cohorts.
- Moore, L., Bartels, T., Persky, D. O., Abraham, I., Kumar, A., & McBride, A. (2021). Outcomes of primary and secondary prophylaxis of chemotherapy-induced and febrile neutropenia in bendamustine plus rituximab regimens in patients with lymphoma and chronic lymphocytic leukemia: real-world, single-center experience. Supportive care in cancer : official journal of the Multinational Association of Supportive Care in Cancer, 29(8), 4867-4874.More infoTo examine the outcomes associated with granulocyte colony stimulating factors (G-CSFs) administered as primary versus secondary prophylaxis in setting of bendamustine plus rituximab (BR) regimens.
- Smith, J., Kumar, A., Stanton, N. A., & Katsanis, E. (2021). Concurrent application of blinatumomab and haploidentical donor leukocyte infusions for refractory primary mediastinal large B-cell lymphoma. Therapeutic advances in hematology, 12, 2040620721994348.More infoPrimary mediastinal large B-cell lymphoma (PMBCL) is a rare hematologic malignancy with distinct clinical and immunopathological features. We report a case of a young male with disease refractory to multiple lines of therapy, including chimeric antigen receptor-T cells, who achieved his first complete remission after haploidentical bone marrow transplantation (haplo-BMT), following donor leukocyte infusions (DLIs) given concurrently with blinatumomab. While DLI has been used after T-replete haplo-BMT with post-transplant cyclophosphamide, there are no reports on its use for PMBCL. Similarly, blinatumomab is active against B-cell lymphomas, but literature is lacking in patients with PMBCL. Our experience illustrates that blinatumomab can be used concurrently with DLI in a haploidentical setting to achieve disease response in PMBCL. Despite our encouraging experience with this case, we would not recommend this approach outside of a clinical trial as blinatumomab may exacerbate the graft host disease risks of DLI, especially in a haploidentical setting. Evaluating this treatment combination in high-risk patients in the setting of a clinical trial may be meaningful.
- Bartels, T., Moore, L., Persky, D. O., Kumar, A., Abraham, I., & McBride, A. (2020). Utilizing a novel four-drug regimen to reduce the incidence of infusion-related reactions for first-dose rituximab infusions: An institutional review of rituximab infusion-related reactions in lymphoma patients.. Journal of Clinical Oncology, 38(15_suppl), e19148-e19148. doi:10.1200/jco.2020.38.15_suppl.e19148More infoe19148 Background: Rituximab is a chimeric monoclonal anti-CD20 antibody utilized in the treatment of several disease states including B-cell non-Hodgkins Lymphoma. Infusion-related reactions (IRR) with rituximab occur with an incidence of up to 77% during the first infusion. The occurrence of an infusion reaction adds to the cost and time of rituximab administrations, especially with prolonged chair time and increased nursing and physician support, and decreased quality of life. The University of Arizona Cancer Center (AZCC) initiated a four-drug premedication regimen consisting of dexamethasone, famotidine, diphenhydramine, and acetaminophen. Our study examined the frequency of first-dose rituximab infusion reactions with our premedication regimen as compared to other published regimens. Methods: A retrospective chart review for all first-dose rituximab infusions was conducted for lymphoma patients treated with BR or RCHOP regimens from 11/2013 through 6/2019 at the AZCC. Data points collected included baseline patient demographics as well as rituximab infusion data. Results: In our study, the median age of the patient population was 64 years (range 22-89), 60.5% were males, and the average BSA was 2 m 2 (range 1.39-3.04). The average rituximab dose was 750 mg (range 521-1140) and the average infusion time was 301 minutes (range 197-512). IRR with first time Rituximab was seen in 19 patients out of 81 total patients (23.5%). Of these IRR, 52.6% occurred in BR versus 47.4% in RCHOP. The most frequent IRR symptoms seen included flushing, itching, feeling hot, and tingling. The average infusion time for IRR patients was 334 minutes versus 284 minutes in patients not experiencing IRR. Grade 2 reactions were reported in 14 patients (73.7%) with grade 3 IRR’s reported in 5 patients (26.3%). Onset of IRR occurred on average during the first 61.6 minutes of the start of infusion (range 10-108 minutes). Conclusions: A novel four drug premedication regimen consisting of corticosteroids, antihistamines H2RA antatagonist and analgesic, resulted in a much lower rituximab infusion reaction rate (23.5%) than what has been previously reported in literature using standard premedications ( > 50%). The utilization of this four drug combination therapy provides the means to reduce first dose rituximab IRR in patients while maximizing patient care and increasing the number of patients who were able to receive Rapid Rituximab during their next cycle of chemotherapy.
- Katsanis, E., Kumar, A., & Smith, J. (2020). Concurrent application of blinatumomab and haploidentical donor leukocyte infusions for refractory primary mediastinal large B-cell lymphoma. Blood online. doi:10.22541/au.159986138.85345714
- Kumar, A., & Persky, D. O. (2020). Treatment of Early (Limited)-Stage DLBCL. Clinical lymphoma, myeloma & leukemia, 20 Suppl 1, S34-S36.
- Kumar, A. (2019). Abstract CT028: Drug-drug interaction potential of EZH2 inhibitor tazemetostat (TAZ). American Association for Cancer Research, 79(13 Supplement), CT028-CT028. doi:10.1158/1538-7445.AM2019-CT028More infoAACR Annual Meeting 2019; March 29-April 3, 2019; Atlanta, GA
- Kumar, A. (2019). Outcomes of Primary and Secondary Prophylaxis of Chemotherapy Induced and Febrile Neutropenia (CIN/FN) in Bendamustine Plus Rituximab (BR) Regimens in Patients with Lymphoma and Chronic Lymphocytic Leukemia (CLL): Real-World, Single-Center Experience. Blood, 134(Supplement_1), 5353.
- Kumar, A. (2019). The efficacy of ibrutinib-based combination therapy for mantle cell lymphoma: A systematic review.. Journal of Clinical Oncology, 37(15_suppl), e19043-e19043.
- Kumar, A., Fraz, M. A., Usman, M., Malik, S. U., Ijaz, A., Durer, C., Durer, S., Tariq, M. J., Khan, A. Y., Qureshi, A., Faridi, W., Nasar, A., & Anwer, F. (2018). Treating Diffuse Large B Cell Lymphoma in the Very Old or Frail Patients. Current treatment options in oncology, 19(10), 50.More infoR-CHOP has been the standard of care for diffuse large B cell lymphoma (DLBCL), curing approximately 60% of patients for more than 2 decades. However, the optimal treatment of patients who are too frail to tolerate this regimen and/or are not candidates for anthracycline therapy continues to be debated. MInT and GELA trials established addition of rituximab to CHOP in DLBCL but excluded patients older than 80 years. Multiple regimens have been tried with varying success in the very elderly, including R-mini-CHOP, R-mini CEOP, R-split CHOP, pre-phase strategies, and R-GCVP. However, there has not been a randomized trial among these strategies. Although addition of novel agents including ibrutinib, brentuximab vedotin, lenalidomide, and many others on the horizon holds promise in this population, none have been tested in a randomized setting or have results awaited. There is also a lack of a validated and easy to use clinical tool in this population to predict patients who will not tolerate R-CHOP. Identifying patients who will not tolerate R-CHOP early with the help of tools like CGA, along with integrating biology-based treatment (ibrutinib, lenalidomide in activated B cell type DLBCL) is being investigated in this population.
- McBride, A., Yiu, K., Elquza, E., Persky, D. O., & Kumar, A. (2018). Transition of dose-adjusted EPOCH therapy into the outpatient healthcare setting: Quality and cost considerations for outpatient treatment.. Journal of Clinical Oncology, 36(30_suppl), 110-110. doi:10.1200/jco.2018.36.30_suppl.110More info110 Background: Dose-adjusted etoposide, prednisone, vincristine, cyclophosphamide, doxorubicin (EPOCH)-containing regimens are frequently utilized in lymphoma, however, outpatient EPOCH or modified inpatient/outpatient EPOCH has not been described extensively. We transitioned inpatient EPOCH to the outpatient setting and modified EPCOH to be given in the inpatient setting with rituximab outpatient to improve quality of care and access to patient assistance programs. We describe our institutional experiences with inpatient and modified EPOCH to the outpatient setting. Methods: A single-center, institutional review board-approved retrospective study was conducted for adults receiving EPOCH-based regimens. Clinical and financial data were collected by chart review for each patient. Descriptive statistics were utilized for analysis. Results: A total of 31 patients received 116 cycles of an EPOCH-containing regimen (11 [9.5%] inpatient), 54 [46.5%] outpatient, and 51 [44.0%] hybrid inpatient and outpatient). Nine outpatient cycles, 9 (17.6%) hybrid cycles and no inpatient cycles resulted in admissions for FN. Two inpatient cycles were delayed (18.2%) due to disease-related procedures and one (9.1%) was delayed due to low blood counts. Five (9.2%) outpatient cycles were delayed due to logistics (i.e. insurance delays, scheduling errors) and two (3.7%) outpatient cycles were delayed due to disease-related adverse events (bowel obstruction, chest pain). Transitioning EPOCH to the outpatient setting decreased overall costs for hospital stays on average by $19,792 per cycle with an overall approximate cost savings to the health-system of 1,114,992 dollars with 432 bed days saved. Costs savings to patients with medications assistance programs is pending final analysis. Conclusions: EPOCH-containing regimens can be safely transitioned into the outpatient setting, side effects can be monitored and outcomes optimized, to better adapt treatment strategies for individualized patient therapies. As new healthcare payment models are developed, outpatient treatments allow for adaptive financial options both for the health-system and the patient.
- Puvvada, S. D., Guillen-Rodriguez, J., Kumar, A., Inclán, L., Heard, K., Rivera, X. I., Anwer, F., Schatz, J. H., Mahadevan, D., & Persky, D. O. (2018). Phase 2 Open-Label Study of Bortezomib, Cladribine, and Rituximab in Advanced, Newly Diagnosed, and Relapsed/Refractory Mantle-Cell and Indolent Lymphomas. Clinical lymphoma, myeloma & leukemia, 18(1), 58-64.More infoMantle-cell lymphoma (MCL) and indolent non-Hodgkin lymphoma (iNHL) are incurable heterogeneous diseases characterized by relapse. There is a need for newer treatments in MCL and iNHL, especially in the relapsed/refractory (R/R) setting. We therefore investigated the novel combination of bortezomib (Velcade), cladribine, and rituximab (VCR) in front-line and R/R settings in MCL and iNHL (NCT00980395).
- Batlevi, C. W., Alperovich, A., Ni, A., Soumerai, J. D., Smith, K., Ying, Z., Caron, P., Drullinsky, P., Gerecitano, J. F., Hamilton, A., Hamlin, P. A., Horwitz, S. M., Kumar, A., Matasar, M. J., Moskowitz, A., Moskowitz, C. H., Noy, A., Palomba, M. L., Portlock, C. S., , Sauter, C., et al. (2017). DEFINING PROGRESSION FREE SURVIVAL AFTER MULTIPLE LINES OF THERAPY AND IMPACT OF DYNAMIC CHANGES IN FLIPI FOR MULTIPLY RELAPSED FOLLICULAR LYMPHOMA IN THE RITUXIMAB ERA. Unknown. doi:10.1002/hon.2438_92
- Naing, A., Heist, R., McClay, E., Richards, D., Mita, M., Kumar, A., Mahadevan, D., & Sundararajan, S. (2017). Phase I single dose, two-period and two-sequence cross-over trial to evaluate the relative bioavailability of two oral pimasertib formulations in advanced cancer patients. Cancer Chemotherapy and Pharmacology.
- Pulluri, B., Kumar, A., Shaheen, M., Jeter, J., & Sundararajan, S. (2017). Tumor microenvironment changes leading to resistance of immune checkpoint inhibitors in metastatic melanoma and strategies to overcome resistance. Pharmacological research, 123, 95-102.More infoImmunotherapy with checkpoint inhibitors targeting CTLA-4 and/or PD-1 receptors independent of the BRAF mutational status and targeted therapy with BRAF and MEK inhibitors in BRAF V600 mutated patients have taken the forefront of advanced melanoma treatment. The main advantage of immunotherapy is its ability to provide durable responses in a subset of patients. However, significant proportions of patients either do not respond or have progression after initial response to immunotherapies. Multiple changes in the tumor microenvironment, such as down regulation of immune checkpoint ligands by tumor, alteration in interferon signaling, and activation of alternate immune suppressive pathways, have been identified as possible reasons for failure of immune checkpoint therapy. Here, we review the resistance mechanisms adopted by cancer cells to checkpoint inhibitor therapy and targeted therapy. In addition, we focus on the available and emerging evidence on tumor microenvironment modulation by BRAF/MEK inhibitor therapy and its role in improving responses to checkpoint inhibitor therapy.
- Kumar, A., & Le, D. T. (2016). Hepatocellular Carcinoma Regression After Cessation of Immunosuppressive Therapy. Journal of clinical oncology : official journal of the American Society of Clinical Oncology, 34(10), e90-2.
- Kumar, A., Sundararajan, S., Puvvada, S., & Persky, D. O. (2016). Limited Stage Aggressive Non-Hodgkin Lymphoma: What Is Optimal Therapy?. Current treatment options in oncology, 17(9), 45.More infoThe seminal SWOG trial S8736 trial established the success of a short course of chemotherapy followed by involved field radiation in treating limited stage aggressive NHL lymphoma. Addition of rituximab offered a surprisingly modest improvement in this disease subset. Radioimmunotherapy could hold a slight advantage over rituximab, but that should be investigated in a randomized trial setting. The role of radiation therapy continues to be widely debated, with interpretation complicated by different trial populations, methods of assessing risk, as well as by differences in timing and dose of radiation. Prolonged course of chemotherapy followed by radiation is certainly not justified in all patients with limited stage disease. Three to four cycles of R-CHOP followed closely by IFRT/ISRT, or six cycles of R-CHOP chemoimmunotherapy (based on the MInT trial) are acceptable options. PET/CT scans may further limit radiation to minority of patients who have residual PET-positive masses. PET/CT-directed treatment strategy is being tested in a US intergroup trial. There is evidence that localized DLBCL has a different biology as compared to advanced stage disease. This relates to propensity of limited stage disease to be proportionately more germinal center B-cell like (GCB) and to have late relapses beyond 5 years. Both biology and imaging need to be integrated in the study of limited stage disease without presumption that it should be approached the same as advanced stage disease.
- Mahadevan, D., Mita, M., Richards, D., McClay, E., Heist, R. S., Kumar, A., Sundararajan, S., & Naing, A. (2017). Phase I single dose, two-period and two-sequence cross-over trial to evaluate the relative bioavailability of two oral pimasertib formulations in advanced cancer patients. Cancer chemotherapy and pharmacology, 79(4), 681-688.More infoA phase I two-period two sequence cross-over study compared the bioavailability of two pimasertib (MSC1936369B/AS703026) formulations (capsule versus tablet) in advanced cancer patients.
- Sundararajan, S., Chen, H., Kumar, A., Tus, K., Yeager, A., & Puvvada, S. (2016). Donor-derived marginal zone lymphoma following reduced-intensity allogeneic peripheral blood stem cell transplant. Leukemia & lymphoma, 57(7), 1735-8.
- Sundararajan, S., Kumar, A., Korde, N., & Agarwal, A. (2016). Smoldering Multiple Myeloma: Emerging Concepts and Therapeutics. Current hematologic malignancy reports, 11(2), 102-10.More infoSmoldering multiple myeloma (SMM) is a pre-malignant condition with an inherent risk for progression to multiple myeloma (MM). The 2014 IMWG guidelines define smoldering multiple myeloma as a monoclonal gammopathy disorder with serum monoclonal protein (IgG or IgA) ≥30 g/L or urinary monoclonal protein ≥500 mg per 24 h and/or clonal bone marrow plasma cells 10-60 % without any myeloma-defining events or amyloidosis. The risk for progression of SMM to MM vary based on clinical, laboratory, imaging, and molecular characteristics. Observation, with periodic monitoring is the current standard of care for SMM. Over last few years, research advances in SMM have led to the delineation of newer risk factors for progression and identification of a "high-risk" group that would potentially benefit from early treatment. This review focuses on advances in the SMM risk-stratification model and recent clinical trials in this patient population.
- Sundararajan, S., Kumar, A., Poongkunran, M., Kannan, A., & Vogelzang, N. J. (2016). Cardiovascular adverse effects of targeted antiangiogenic drugs: mechanisms and management. Future oncology (London, England), 12(8), 1067-80.More infoAnticancer treatment has evolved enormously over the last decade. Drugs targeting receptor tyrosine kinases, VEGFR and EGFR have changed the treatment landscape of certain cancers and have shifted the theme of anticancer therapy toward personalized care. However, these newer agents also come with unique side-effect profiles not seen with conventional chemotherapy including serious cardiovascular adverse effects. Hence, meticulous understanding of the adverse effects is crucial in maximizing clinical benefits and minimizing detrimental effects of these newer drugs. We have reviewed the cardiovascular adverse effects of anti-VEGF therapy in this article.
- Kumar, A., & Puvvada, S. (2015). Mantle cell lymphoma presenting as cardiac tamponade. Blood, 126(10), 1255.
- Kumar, A., Dagar, M., Herman, J., Iacobuzio-Donahue, C., & Laheru, D. (2015). CNS involvement in pancreatic adenocarcinoma: a report of eight cases from the Johns Hopkins Hospital and review of literature. Journal of gastrointestinal cancer, 46(1), 5-8.More infoCNS metastasis of pancreatic cancer is extremely rare, although systemic metastasis is very common. We present eight such cases with various forms of nervous system involvement.
- Sundararajan, S., Chen, H., Kumar, A., Tus, K., Yeager, A. M., & Puvvada, S. D. (2015). Donor-derived marginal zone lymphoma following reduced-intensity allogeneic peripheral blood stem cell transplant. Clinical Leukemia and Lymphoma. doi:10.3109/10428194.2015.1113282More infoPost-transplant lympho-proliferative disorders (PTLD) pose significant therapeutic challenges accompanied by increase in morbidity and mortality. The timing and severity of PTLD is often variable; ...
- Kumar, A., Dagar, M., Herman, J. M., Iacobuzio–Donahue, C. A., & Laheru, D. (2014). CNS Involvement in Pancreatic Adenocarcinoma: a Report of Eight Cases from the Johns Hopkins Hospital and Review of Literature. Journal of GI cancer. doi:10.1007/s12029-014-9667-yMore infoCNS metastasis of pancreatic cancer is extremely rare, although systemic metastasis is very common. We present eight such cases with various forms of nervous system involvement. Data was gathered from chart review of 800 patients with pancreatic cancer treated between 2004 and 2012 of which eight patients are described with CNS metastases. The median age of patients was 61.5 years and the median time to develop CNS metastasis was 29 months. Interestingly, two patients had no other sites of metastasis. The treatment modalities tried included resection followed by radiation, resection alone, or whole brain radiation.
Presentations
- Kumar, A. (2022, July/Summer). Lymphoma. ASCO Direct Hematological Malignancies. Las Vegas, NV: ASCO.
- Kumar, A. (2022, November/Winter). Critical Reviews. Introduction to Clinical and Translational Research "Fellows Boot Camp". Tucson, AZ: UACC.
- Kumar, A. (2020, October/Fall). Lymphoma 101 for Newly Diagnosed Patients/Caregivers. 25th annual North American Educational Forum on Lymphoma / Lymphoma Foundation. virtual.
- Kumar, A. (2020, October/Fall). Lymphoma. PRIME: Oncology Program. virtual: PRIME.
- Kumar, A. (2016, December/Winter). A Phase II, Open-Label Study of Bortezomib (Velcade), Cladribine and Rituximab (VCR) in Advanced, Newly Diagnosed and Relapsed/Refractory Mantle Cell and Indolent Lymphomas. American Society of Hematology (ASH) Annual Meeting. San Diego, CA: ASH.
Poster Presentations
- Abraham, I. L., Kumar, A., Persky, D. O., & Anwar, F. (2017, December/Winter). 4678 Economic Evaluation for the US of Venetoclax Versus Allogeneic Hematopoietic Stem-Cell Transplantation for Patients with Relapsed or Refractory Chronic Lymphocytic Leukemia with 17p Deletion. ASH Annual Meeting. Atllanta, GA: ASH.
- Abraham, I. L., Kumar, A., Persky, D. O., & Anwar, F. (2017, December/Winter). 4679 Economic Evaluation for the US of Ibrutinib Versus Allogeneic Hematopoietic Stem-Cell Transplantation for Patients with Relapsed or Refractory Chronic Lymphocytic Leukemia with 17p Deletion. ASH Annual Conference. Atlanta GA: ASH.
- Kumar, A. (2016, January/Winter). Epidemiology and survival of metastatic gastrointestinal stromal tumor: A Surveillance, Epidemiology, and End Results (SEER) database review. GI ASCO Symposium. San Francisco, CA: ASCO.
- Kumar, A. (2016, January/Winter). Epidemiology and survival of small cell carcinoma of gastrointestinal tract: A Surveillance, Epidemiology, and End Results (SEER) database review. GI ASCO Symposium. San Francisco, CA: ASCO.
- Kumar, A. (2014, April/Spring). Retrospective Analysis of CML treated at St. Agnes Hospital. ACP. Orlando, FL: American College of Physicians.
- Kumar, A. (2012, April/Spring). Miliary BCG Pneumonitis: A Rare Complication of a common therapy. ACP Annual Meeting. New Orleans, Louisiana: ACP.
- Kumar, A. (2012, April/Spring). Systemic mastocytosis: The mystery of a 89 year old with chronic loose stools.. ACP Annual Meeting. New Orleans, Louisiana: ACP.
Others
- Rivera, X., Ayala, A., Inclan, L., Schatz, J., Guillen-Rodriguez, J., Anwer, F., Kumar, A., Puvvada, S. D., Persky, D. O., Mahadevan, D., & Sundararajan, S. (2016, December). A phase II, open-label study of bortezomib (Velcade®), cladribine, and rituximab (VCR) in advanced, newly diagnosed and relapsed /refractory mantle Cell and indolent Lymphomas.