Try a new search

Format these results:

Searched for:

person:hsuj05

in-biosketch:true

Total Results:

33


XPO1 Enables Adaptive Regulation of mRNA Export Required for Genotoxic Stress Tolerance in Cancer Cells

Marullo, Rossella; Rutherford, Sarah C; Revuelta, Maria V; Zamponi, Nahuel; Culjkovic-Kraljacic, Biljana; Kotlov, Nikita; Di Siervi, Nicolás; Lara-Garcia, Juan; Allan, John N; Ruan, Jia; Furman, Richard R; Chen, Zhengming; Shore, Tsiporah B; Phillips, Adrienne A; Mayer, Sebastian; Hsu, Jingmei; van Besien, Koen; Leonard, John P; Borden, Katherine L B; Inghirami, Giorgio; Martin, Peter; Cerchietti, Leandro
UNLABELLED:Exportin-1 (XPO1), the main soluble nuclear export receptor in eukaryotic cells, is frequently overexpressed in diffuse large B-cell lymphoma (DLBCL). A selective XPO1 inhibitor, selinexor, received approval as single agent for relapsed or refractory (R/R) DLBCL. Elucidating the mechanisms by which XPO1 overexpression supports cancer cells could facilitate further clinical development of XPO1 inhibitors. We uncovered here that XPO1 overexpression increases tolerance to genotoxic stress, leading to a poor response to chemoimmunotherapy. Upon DNA damage induced by MYC expression or exogenous compounds, XPO1 bound and exported EIF4E and THOC4 carrying DNA damage repair mRNAs, thereby increasing synthesis of DNA damage repair proteins under conditions of increased turnover. Consequently, XPO1 inhibition decreased the capacity of lymphoma cells to repair DNA damage and ultimately resulted in increased cytotoxicity. In a phase I clinical trial conducted in R/R DLBCL, the combination of selinexor with second-line chemoimmunotherapy was tolerated with early indication of efficacy. Overall, this study reveals that XPO1 overexpression plays a critical role in the increased tolerance of cancer cells to DNA damage while providing new insights to optimize the clinical development of XPO1 inhibitors. SIGNIFICANCE/UNASSIGNED:XPO1 regulates the dynamic ribonucleoprotein nuclear export in response to genotoxic stress to support tolerance and can be targeted to enhance the sensitivity of cancer cells to endogenous and exogenous DNA damage. See related commentary by Knittel and Reinhardt, p. 3.
PMCID:10758694
PMID: 37801604
ISSN: 1538-7445
CID: 5625742

PTCy, Abatacept, and Short Course of Tacrolimus for GvHD Prevention Following Haploidentical Transplantation

Al-Homsi, A Samer; Cirrone, Frank; Wo, Stephanie; Cole, Kelli; Suarez-Londono, J Andres; Gardner, Sharon L; Hsu, Jingmei; Stocker, Kelsey; Bruno, Benedetto; Goldberg, Judith D; Levinson, Benjamin A; Abdul-Hay, Maher
Reducing the incidence of graft-versus host disease (GvHD) following haploidentical hematopoietic stem cell transplantation (HSCT) is warranted. Post-transplant cyclophosphamide (PTCy) is the main agent used for GvHD prevention in this setting. It remains unknown if costimulation blockade can be safely combined with PTCy and enhance its efficacy. We performed a phase Ib-II clinical trial to examine the combination of PTCy, abatacept and a short course of tacrolimus (CAST) following peripheral blood haploidentical HSCT. The primary end-point was the incidence of acute GvHD grades II-IV at day +120. The study enrolled 46 patients with a median age of 60 years (range: 18 to 74). The cumulative incidence of acute GvHD grades II-IV and III-IV was 17.4% (95% CI, 9.2% to 32.9%) and 4.4% (95% CI, 1.1% to 17.1%). With a median follow-up of 15.3 months, the cumulative incidence of one-year treatment-related mortality is 4.4% (95% CI, 1.1% to 17.1%). The estimated one-year chronic GvHD moderate to severe rate, relapse rate, progression-free survival, overall survival, and GvHD- and relapse-free survival were 15.9% (95% CI, 8% to 31.7%), 11.7% (95% CI, 5% to 27.2%), 84.1% (95% CI, 73.8% to 95.7%), 85.9% (95% CI, 75.9% to 97.2%) and 66.1% (95% CI, 53.4% to 81.8%), respectively. Toxicities were similar to those expected in patients receiving haploidentical HSCT. This clinical trial showed that CAST regimen is safe and effective in reducing the rate of grades II-IV acute GvHD following haploidentical peripheral blood HSCT (NCT04503616 at https://clinicaltrials.gov/ct2/show/NCT04503616).
PMID: 37163349
ISSN: 2473-9537
CID: 5509352

HIV-1 remission and possible cure in a woman after haplo-cord blood transplant

Hsu, Jingmei; Van Besien, Koen; Glesby, Marshall J; Pahwa, Savita; Coletti, Anne; Warshaw, Meredith G; Petz, Larry; Moore, Theodore B; Chen, Ya Hui; Pallikkuth, Suresh; Dhummakupt, Adit; Cortado, Ruth; Golner, Amanda; Bone, Frederic; Baldo, Maria; Riches, Marcie; Mellors, John W; Tobin, Nicole H; Browning, Renee; Persaud, Deborah; Bryson, Yvonne
Previously, two men were cured of HIV-1 through CCR5Δ32 homozygous (CCR5Δ32/Δ32) allogeneic adult stem cell transplant. We report the first remission and possible HIV-1 cure in a mixed-race woman who received a CCR5Δ32/Δ32 haplo-cord transplant (cord blood cells combined with haploidentical stem cells from an adult) to treat acute myeloid leukemia (AML). Peripheral blood chimerism was 100% CCR5Δ32/Δ32 cord blood by week 14 post-transplant and persisted through 4.8 years of follow-up. Immune reconstitution was associated with (1) loss of detectable replication-competent HIV-1 reservoirs, (2) loss of HIV-1-specific immune responses, (3) in vitro resistance to X4 and R5 laboratory variants, including pre-transplant autologous latent reservoir isolates, and (4) 18 months of HIV-1 control with aviremia, off antiretroviral therapy, starting at 37 months post-transplant. CCR5Δ32/Δ32 haplo-cord transplant achieved remission and a possible HIV-1 cure for a person of diverse ancestry, living with HIV-1, who required a stem cell transplant for acute leukemia.
PMID: 36931242
ISSN: 1097-4172
CID: 5468462

[S.l.] : Tandem Meetings, Transplantation & Cellular Therapy Meetings of ASTCT and CIBMTR, 2023

Phase Ib-II Study of Post-Transplant Cyclophosphamide, Abatacept and Short Course of Tacrolimus (CAST) for Graft-Versus-Host Disease Prevention Following Haploidentical Peripheral Blood Stem Cell Transplantation

Al-Homsi, A Samer; Cirrone, Frank; Cole, Kelli; Londono, J Andres-Suarez; Gardner, Sharon; Hsu, Jingmei; Wo, Stephanie; Stocker, Kelsey; Goldberg, Judith; Levinson, Benjamin; Abdul-Hay, Maher
(Website)
CID: 5515802

Reply to Caldwell et al

Satlin, Michael J; Chen, Liang; Douglass, Claire; Hovan, Michael; Davidson, Emily; Soave, Rosemary; La Spina, Marisa; Gomez-Arteaga, Alexandra; van Besien, Koen; Mayer, Sebastian; Phillips, Adrienne; Hsu, Jing-Mei; Malherbe, Rianna; Small, Catherine B; Jenkins, Stephen G; Westblade, Lars F; Kreiswirth, Barry N; Walsh, Thomas J
PMID: 34864913
ISSN: 1537-6591
CID: 5204022

Axicabtagene Ciloleucel in Patients Ineligible for ZUMA-1 Because of CNS Involvement and/or HIV: A Multicenter Experience

Yuen, Carlen A; Hsu, Jing-Mei; Van Besien, Koen; Reshef, Ran; Iwamoto, Fabio M; Haggiagi, Aya; Liechty, Benjamin; Zhang, Cenai; Wesley, Sarah F; Magge, Rajiv
Secondary central nervous system lymphoma (SCNSL) is associated with poor prognosis and new therapeutic approaches are needed. The pivotal trial that led to US Food and Drug Administration (FDA) approval of axicabtagene ciloleucel excluded patients with SCNSL and human immunodeficiency virus. In this multi-institutional retrospective study, 14 SCNSL patients treated with axicabtagene ciloleucel, 3 of whom had human immunodeficiency virus, experienced rates of severe neurotoxicity and complete response of 32% and 58%, respectively. This is similar to rates observed in the pivotal ZUMA-1 trial that led to the approval of axi-cel at median follow-up of 5.9 months. Chimeric antigen receptor T-cell therapy is potentially a life-saving therapy for SCNSL patients and should not be withheld.
PMID: 35404315
ISSN: 1537-4513
CID: 5204032

Post-Transplant Cyclophosphamide, Abatacept and Short Course of Tacrolimus (CAST) for Graft-Versus-Host Disease Prevention Following Haploidentical Peripheral Blood Stem Cell Transplantation [Meeting Abstract]

Al-Homsi, A. Samer; Cirrone, Frank; Cole, Kelli; Suarez-Londono, Jaime Andres; Gardner, Sharon L.; Hsu, Jingmei; Wo, Stephanie; Stocker, Kelsey; Bruno, Benedetto; Goldberg, Judith; Levinson, Benjamin; Abdul-Hay, Maher
ISI:000893230300285
ISSN: 0006-4971
CID: 5515762

Planned Granulocyte Colony-Stimulating Factor Adversely Impacts Survival after Allogeneic Hematopoietic Cell Transplantation Performed with Thymoglobulin for Myeloid Malignancy

Orfali, Nina; Zhang, Mei-Jie; Allbee-Johnson, Mariam; Boelens, Jaap Jan; Artz, Andrew S; Brunstein, Claudio G; McNiece, Ian K; Milano, Filippo; Abid, Muhammad Bilal; Chee, Lynette; Diaz, Miguel A; Grunwald, Michael R; Hematti, Peiman; Hsu, Jingmei; Lazarus, Hillard M; Munshi, Pashna N; Prestidge, Timothy; Ringden, Olle; Rizzieri, David; Riches, Marcie L; Seo, Sachiko; Solh, Melhem; Solomon, Scott; Szwajcer, David; Yared, Jean; van Besien, Koen; Eapen, Mary
The in vivo depletion of recipient and donor T lymphocytes using antithymocyte globulin (ATG; Thymoglobulin) is widely adopted in allogeneic hematopoietic stem cell transplantation (HCT) to reduce the incidence of both graft failure and graft-versus-host disease (GVHD). However, excess toxicity to donor lymphocytes may hamper immune reconstitution, compromising antitumor effects and increasing infection. Granulocyte-colony stimulating factor (G-CSF) administered early after HCT may increase ATG-mediated lymphotoxicity. This study aimed to investigate the effect of an interaction between ATG and post-transplantation granulocyte colony-stimulating factor (G-CSF) on allogeneic HCT outcomes, using the Center for International Blood and Marrow Transplant Research (CIBMTR) registry. We studied patients age ≥18 years with acute myelogenous leukemia (AML) and myelodysplastic syndrome (MDS) who received Thymoglobulin-containing preparative regimens for HLA-matched sibling/unrelated or mismatched unrelated donor HCT between 2010 and 2018. The effect of planned G-CSF that was started between pretransplantation day 3 and post-transplantation day 12 was studied in comparison with transplantations that did not include G-CSF. Cox regression models were built to identify risk factors associated with outcomes at 1 year after transplantation. A total of 874 patients met the study eligibility criteria, of whom 459 (53%) received planned G-CSF. HCT with planned G-CSF was associated with a significantly increased risk for nonrelapse mortality (NRM) (hazard ratio [HR] 2.03; P <.0001; 21% versus 12%) compared to HCT without G-CSF. The 6-month incidence of viral infection was higher with G-CSF (56% versus 47%; P = .007), with a particular increase in Epstein-Barr virus infections (19% versus 11%; P = .002). The observed higher NRM with planned G-CSF led to lower overall survival (HR, 1.52; P = .0005; 61% versus 72%). There was no difference in GVHD risk between the treatment groups. We performed 2 subgroup analyses showing that our findings held true in patients age ≥50 years and in centers where G-CSF was used in some, but not all, patients. In allogeneic peripheral blood HCT performed with Thymoglobulin for AML and MDS, G-CSF administered early post-transplantation resulted in a 2-fold increase in NRM and a 10% absolute decrement in survival. The use of planned G-CSF in the early post-transplantation period should be carefully considered on an individual patient basis, weighing any perceived benefits against these risks.
PMCID:8671234
PMID: 34507002
ISSN: 2666-6367
CID: 5204002

Long-term clinical outcomes of tisagenlecleucel in patients with relapsed or refractory aggressive B-cell lymphomas (JULIET): a multicentre, open-label, single-arm, phase 2 study

Schuster, Stephen J; Tam, Constantine S; Borchmann, Peter; Worel, Nina; McGuirk, Joseph P; Holte, Harald; Waller, Edmund K; Jaglowski, Samantha; Bishop, Michael R; Damon, Lloyd E; Foley, Stephen Ronan; Westin, Jason R; Fleury, Isabelle; Ho, P Joy; Mielke, Stephan; Teshima, Takanori; Janakiram, Murali; Hsu, Jing-Mei; Izutsu, Koji; Kersten, Marie José; Ghosh, Monalisa; Wagner-Johnston, Nina; Kato, Koji; Corradini, Paolo; Martinez-Prieto, Marcela; Han, Xia; Tiwari, Ranjan; Salles, Gilles; Maziarz, Richard T
BACKGROUND:In the primary analysis of the pivotal JULIET trial of tisagenlecleucel, an autologous anti-CD19 chimeric antigen receptor (CAR) T-cell therapy, the best overall response rate was 52% and the complete response rate was 40% in 93 evaluable adult patients with relapsed or refractory aggressive B-cell lymphomas. We aimed to do a long-term follow-up analysis of the clinical outcomes and correlative analyses of activity and safety in the full adult cohort. METHODS:viable transduced CAR T cells). The primary endpoint was overall response rate (ie, the proportion of patients with a best overall disease response of a complete response or partial response using the Lugano classification, as assessed by an independent review committee) at any time post-infusion and was analysed in all patients who received tisagenlecleucel (the full analysis set). Safety was analysed in all patients who received tisagenlecleucel. JULIET is registered with ClinialTrials.gov, NCT02445248, and is ongoing. FINDINGS:Between July 29, 2015, and Nov 2, 2017, 167 patients were enrolled. As of Feb 20, 2020, 115 patients had received tisagenlecleucel infusion and were included in the full analysis set. At a median follow-up of 40·3 months (IQR 37·8-43·8), the overall response rate was 53·0% (95% CI 43·5-62·4; 61 of 115 patients), with 45 (39%) patients having a complete response as their best overall response. The most common grade 3-4 adverse events were anaemia (45 [39%]), decreased neutrophil count (39 [34%]), decreased white blood cell count (37 [32%]), decreased platelet count (32 [28%]), cytokine release syndrome (26 [23%]), neutropenia (23 [20%]), febrile neutropenia (19 [17%]), hypophosphataemia (15 [13%]), and thrombocytopenia (14 [12%]). The most common treatment-related serious adverse events were cytokine release syndrome (31 [27%]), febrile neutropenia (seven [6%]), pyrexia (six [5%]), pancytopenia (three [3%]), and pneumonia (three [3%]). No treatment-related deaths were reported. INTERPRETATION:Tisagenlecleucel shows durable activity and manageable safety profiles in adult patients with relapsed or refractory aggressive B-cell lymphomas. For patients with large B-cell lymphomas that are refractory to chemoimmunotherapy or relapsing after second-line therapies, tisagenlecleucel compares favourably with respect to risk-benefit relative to conventional therapeutic approaches (eg, salvage chemotherapy). FUNDING:Novartis Pharmaceuticals.
PMID: 34516954
ISSN: 1474-5488
CID: 5204012

Sequential intensive chemotherapy followed by autologous or allogeneic transplantation for refractory lymphoma

Orfali, Nina; Jhanwar, Yuliya; Koo, Calvin; Pasciolla, Michelle; Baldo, Maria; Cuvilly, Edwidge; Furman, Richard; Gergis, Usama; Greenberg, June; Guarneri, Danielle; Hsu, Jing-Mei; Leonard, John P; Mark, Tomer; Mayer, Sebastian; Maignan, Kathleen; Martin, Peter; Opong, Adomah; Pearse, Roger; Phillips, Adrienne; Rossi, Adriana; Ruan, Jia; Rutherford, Sarah C; Ryan, Jessy; Suhu, Grace; Van Besien, Koen; Shore, Tsiporah
We evaluate the safety of bendamustine as a bridge to stem cell transplantation (SCT) in patients with relapsed/refractory lymphoma and residual disease after salvage therapy. Thirty-four subjects without complete responses (CR) received bendamustine 200 mg/m2/day for 2 days followed 14 days later by SCT. Sixteen subjects in partial remission (PR) with maximal FDG-PET SUVs ≤8 prior to bendamustine received autologous SCT, while 13 with suboptimal responses were allografted. Five subjects did not proceed to transplant. No bendamustine toxicities precluded transplantation and no detrimental effect on engraftment or early treatment-related mortality (TRM) was attributable to bendamustine. At 1 year, 75% of auto-recipients and 31% of allo-recipients were alive with CR. Two subjects in the autologous arm developed therapy-related myeloid neoplasia (t-MN). In conclusion, a bendamustine bridge to SCT can be administered without early toxicity to patients with suboptimal responses to salvage chemotherapy. However this approach may increase the risk of t-MN. (NCT02059239).Supplemental data for this article is available online at here.
PMID: 33586581
ISSN: 1029-2403
CID: 5203992