Clinical Focus


  • Blood and Marrow Transplantation, Pediatric
  • Cell and Gene therapy
  • Pediatric Hematology-Oncology

Academic Appointments


Administrative Appointments


  • Clinical Director, Pediatric Stem Cell Transplant (2008 - Present)
  • Section chief, Pediatric Stem Cell Transplantation (2010 - Present)

Honors & Awards


  • Fanconi Anemia Appreciation award, Fanconi Anemia Research Fund (October 2023)
  • DISTINGUISHED SERVICE AWARD, STANFORD CHILDREN'S HEALTH/LUCILE PACKARD CHILDRENS HOSPITAL (MAY14, 2018)
  • President's award, Best Woman Candidate in India in Medical School Examinations (1979)

Professional Education


  • Residency: Cincinnati Children's Hospital Medical Center Pediatric Residency (1995) OH
  • Residency: MGM Medical College (1983) India
  • Medical Education: MGM Medical College (1983) India
  • Internship: MGM Medical College (1981) India
  • MD, Indore University, Pediatrics (1983)
  • M.B.B.S, Indore University (1980)

Community and International Work


  • Crescent Zakat Fund, Children's hospital at Stanford

    Topic

    A philanthropic fund to support the Muslim families

    Populations Served

    Muslim families coming from outside the bay area

    Location

    Bay Area

    Ongoing Project

    Yes

    Opportunities for Student Involvement

    Yes

  • Binns Program for Cord Blood Research, Children's hospital at Stanford

    Topic

    Collection of umbilical cord blood for biomedical research

    Populations Served

    All

    Location

    Bay Area

    Ongoing Project

    Yes

    Opportunities for Student Involvement

    Yes

  • volunteer donor recruitment for bone marrow transplants

    Topic

    increasing the minority participation in the unrelated volunteer donor pool

    Partnering Organization(s)

    NMDP

    Populations Served

    Indian subcontinent

    Location

    Bay Area

    Ongoing Project

    No

    Opportunities for Student Involvement

    No

Current Research and Scholarly Interests


I am a highly trained Pediatric stem cell transplant physician with a strong basic science background and experience with translational research. I trained extensively in India and premier institutions in USA as a pediatric Hematologist-Oncologist and transplant physician. My experience in this field also includes extensive expertise in development of stem cell assays and in vivo models of human hematopoiesis .
I started the clinical Umbilical cord blood transplant program at Cincinnati Children's. Through my research efforts we were able to develop a sterile system for collection and use of cord blood cells. This endeavor later contributed in establishing methods to collect and store cord blood for clinical use. In the laboratory we were able to set up the assays to identify and collect highly purified hematopoietic cells from the cord blood. The engraftment and expansion potential of the cord blood derived hematopoietic cells was studied in the immune deficient mice. These models were then used to develop assays for gene transfer in Fanconi Anemia.
Currently, I have been at Stanford university and Children's hospital at Stanford for the past 23 years and served as the medical director and section chief of Pediatric Stem Cell Transplant program until 2019.
My focus is entirely clinical and translational research to reduce toxicity from high doses of conditioning chemotherapy and radiation therapy, cellular therapies to reduce graft vs host disease, graft manipulation to reduce complications from graft vs. host disease in patients who receive mismatched donor stem cells and gene therapy for genetic disorders like IPEX and Fanconi Anemia.
I have expertise in phase I clinical trial design and implementation in cell and gene therapies. I have written, implemented and lead 6 phase 1 clinical trials at Stanford in the past 8 years.
My most innovative clinical Trial is reducing toxicity from chemotherapy during conditioning for stem cell transplants, where we have a phase 1/2 clinical trial using CKIT antibody (JSP 191) successfully in patients with SCID and Fanconi Anemia.
Recently we obtained a CLIN 2 grant from CIRM for using the JSP 191 antibody in patients with Fanconi Anemia.
My goal is to keep pursuing safety in stem cell transplantation for children who have life threatening disorders and keep pushing the envelop to save the precious lives.

Clinical Trials


  • CD4^LVFOXP3 in Participants With IPEX Recruiting

    This first-in-human, Phase 1 clinical trial will test the feasibility of the manufacturing and the safety of the administration of CD4^LVFOXP3 in up to 36 evaluable human participants with IPEX and evaluate the impact of the CD4^LVFOXP3 infusion on the disease.

    View full details

  • Depleted Donor Stem Cell Transplant in Children and Adults With Fanconi Anemia After Being Conditioned With a Regimen Containing Briquilimab Recruiting

    The objective of this clinical trial is to develop a cell therapy for Fanconi Anemia which enables enhanced donor hematopoietic and immune reconstitution with decreased toxicity by transplanting depleted stem cells from a donor after using an experimental antibody treatment called JSP-191 as a part of conditioning. This experimental treatment will hopefully cause fewer side effects than chemotherapy (the current standard of care method). Participants will be administered the conditioning regimen, are assessed until they receive the depleted stem cell infusion, and will be followed for up to 2 years after the cell infusion.

    View full details

  • Long Term Effects On Recipients of Hematopoietic Stem Cell Transplantation Recruiting

    This project allows for the systematic collection and analysis of long-term follow-up clinical parameters in children who have received a stem cell transplant. The data collected will assist in determining appropriate intervention and treatment plans for patients enrolled on this study. In addition, future patients may benefit by having the ability to anticipate problems and develop methods of prevention or early intervention.

    View full details

  • Stem Cell Transplant From Donors After Alpha Beta Cell Depletion in Children and Adults With T-allo10 Cells Addback Recruiting

    The purpose of this study is to determine the safety of a cell therapy, T-allo10, after αβdepleted-HSCT in the hopes that it will boost the adaptive immune reconstitution of the patient while sparing the risk of developing severe Graft-versus-Host Disease (GvHD). The primary objective of Phase 1a is to determine the recommended Phase 2 dose (RP2D) administered after infusion of αβdepleted-HSCT in children and young adults with hematologic malignancies. A Phase 1b extension will occur after dose escalation, enrolling at the RP2D for the T-allo10 cells determined in the Phase 1 portion to evaluate the safety and efficacy of infusion of T-allo10 after receipt of αβdepleted-HSCT. Additionally, Phase 1b aims to explore improvements in immune reconstitution. All participants on this study must be enrolled on another study: NCT04249830

    View full details

  • Use of T-allo10 in Hematopoietic Stem Cell Transplantation (HSCT) for Blood Disorders Recruiting

    A significant number of patients with hematologic malignancies need a hematopoietic stem cell transplant (HSCT) to be cured. Only about 50% of these patients have a fully matched donor, the remaining patients will require an HSCT from a mismatched related or unrelated donor. Almost 60% of these mismatched donor HSCTs will result in graft-versus-host disease (GvHD), which can cause significant morbidity and increased non-relapse mortality. GvHD is caused by the donor effector T cells present in the HSC graft that recognize and react against the mismatched patient's tissues. Researchers and physicians at Lucile Packard Children's Hospital, Stanford are working to prevent GvHD after HSCT with a new clinical trial. The objective of this clinical program is to develop a cell therapy to prevent GvHD and induce graft tolerance in patients receiving mismatched unmanipulated donor HSCT. The cell therapy consists of a cell preparation from the same donor of the HSCT (T-allo10) containing T regulatory type 1 (Tr1) cells able to suppress allogenic (host-specific) responses, thus decreasing the incidence of GvHD. This is the first trial of its kind in pediatric patients and is only available at Lucile Packard Children's Hospital, Stanford. The purpose of this phase 1 study is to determine the safety and tolerability of a cell therapy, T-allo10, to prevent GvHD in patients receiving mismatched related or mismatched unrelated unmanipulated donor HSCT for hematologic malignancies.

    View full details

  • Donor Umbilical Cord Blood Transplant With or Without Ex-vivo Expanded Cord Blood Progenitor Cells in Treating Patients With Acute Myeloid Leukemia, Acute Lymphoblastic Leukemia, Chronic Myelogenous Leukemia, or Myelodysplastic Syndromes Not Recruiting

    This randomized phase II trial studies how well donor umbilical cord blood transplant with or without ex-vivo expanded cord blood progenitor cells works in treating patients with acute myeloid leukemia, acute lymphoblastic leukemia, chronic myelogenous leukemia, or myelodysplastic syndromes. Giving chemotherapy and total-body irradiation before a donor umbilical cord blood transplant helps stop the growth of cancer cells. It may also stop the patient's immune system from rejecting the donor's cells. When the healthy stem cells and ex-vivo expanded cord blood progenitor cells are infused into the patient they may help the patient's bone marrow make stem cells, red blood cells, white blood cells, and platelets. It is not yet known whether giving donor umbilical cord blood transplant plus ex-vivo expanded cord blood progenitor cells is more effective than giving a donor umbilical cord blood transplant alone.

    Stanford is currently not accepting patients for this trial. For more information, please contact Physician Referrals, 650-723-0822.

    View full details

  • Expanded Access Protocol to Provide Brincidofovir for the Treatment of Serious Adenovirus Infection or Disease Not Recruiting

    Provide patients with serious AdV infection or disease access to treatment with BCV.

    Stanford is currently not accepting patients for this trial.

    View full details

  • Safety Study of Gene Modified Donor T-cells Following TCRαβ+ Depleted Stem Cell Transplant Not Recruiting

    This study will evaluate pediatric patients with malignant or non-malignant blood cell disorders who are having a blood stem cell transplant depleted of T cell receptor (TCR) alfa and beta cells that comes from a partially matched family donor. The study will assess whether immune cells, called T cells, from the family donor, that are specially grown in the laboratory and given back to the patient along with the stem cell transplant can help the immune system recover faster after transplant. As a safety measure these T cells have been programmed with a self-destruct switch so that they can be destroyed if they start to react against tissues (Graft versus host disease).

    Stanford is currently not accepting patients for this trial. For more information, please contact Cancer Clinical Trials Office (CCTO), 650-498-7061.

    View full details

  • Sirolimus and Mycophenolate Mofetil in Preventing GVHD in Patients With Hematologic Malignancies Undergoing HSCT Not Recruiting

    This pilot phase I/II trial studies the side effects and how well sirolimus and mycophenolate mofetil work in preventing graft versus host disease (GvHD) in patients with hematologic malignancies undergoing hematopoietic stem cell transplant (HSCT). Biological therapies, such as sirolimus and mycophenolate mofetil, use substances made from living organisms that may stimulate or suppress the immune system in different ways and stop tumor cells from growing. Giving sirolimus and mycophenolate mofetil after hematopoietic stem cell transplant may be better in preventing graft-versus-host disease.

    Stanford is currently not accepting patients for this trial. For more information, please contact Gopin Saini, 650-725-9032.

    View full details

  • Study to Assess Brincidofovir Treatment of Serious Diseases or Conditions Caused by Double-stranded DNA Viruses Not Recruiting

    This was a multicenter, open-label study of oral brincidofovir (BCV) treatment of serious disease or conditions caused by double-stranded DNA (dsDNA) virus(es). Subjects received either a weight-based or a fixed dose of oral BCV once weekly (QW) or twice weekly (BIW) for up to 3 months until clinical disease was resolved or stabilized and/or viral DNA by polymerase chain reaction testing was negative for 4 consecutive weeks, whichever was longer. Under the first protocol amendment, adults and adolescents (≥13 years) received 200 mg or 300 mg BCV BIW (not to exceed 4 mg/kg total weekly dose) depending on the difficulty of treating their disease (i.e., Group 1 or Group 2, respectively), and pediatric subjects (≤12 years) received 4 mg/kg BCV BIW. Under the second protocol amendment, adults and adolescents (≥13 years), regardless of viral infection/disease, had a maximum weekly dose of 200 mg, i.e., 200 mg QW or 100 mg BIW; not to exceed 4mg/kg total weekly dose. Pediatric subjects (≤12 years), regardless of viral infection/disease, had a maximum weekly dose of 4 mg/kg, i.e., 4 mg/kg QW or 2 mg/kg BIW; not to exceed 200 mg.

    Stanford is currently not accepting patients for this trial. For more information, please contact Julia Buckingham, (650) 736 - 1556.

    View full details

2023-24 Courses


All Publications


  • Antiviral cellular therapy for enhancing T-cell reconstitution before or after hematopoietic stem cell transplantation (ACES): a two-arm, open label phase II interventional trial of pediatric patients with risk factor assessment. Nature communications Keller, M. D., Hanley, P. J., Chi, Y., Aguayo-Hiraldo, P., Dvorak, C. C., Verneris, M. R., Kohn, D. B., Pai, S., Davila Saldana, B. J., Hanisch, B., Quigg, T. C., Adams, R. H., Dahlberg, A., Chandrakasan, S., Hasan, H., Malvar, J., Jensen-Wachspress, M. A., Lazarski, C. A., Sani, G., Idso, J. M., Lang, H., Chansky, P., McCann, C. D., Tanna, J., Abraham, A. A., Webb, J. L., Shibli, A., Keating, A. K., Satwani, P., Muranski, P., Hall, E., Eckrich, M. J., Shereck, E., Miller, H., Mamcarz, E., Agarwal, R., De Oliveira, S. N., Vander Lugt, M. T., Ebens, C. L., Aquino, V. M., Bednarski, J. J., Chu, J., Parikh, S., Whangbo, J., Lionakis, M., Zambidis, E. T., Gourdine, E., Bollard, C. M., Pulsipher, M. A. 2024; 15 (1): 3258

    Abstract

    Viral infections remain a major risk in immunocompromised pediatric patients, and virus-specific T cell (VST) therapy has been successful for treatment of refractory viral infections in prior studies. We performed a phase II multicenter study (NCT03475212) for the treatment of pediatric patients with inborn errors of immunity and/or post allogeneic hematopoietic stem cell transplant with refractory viral infections using partially-HLA matched VSTs targeting cytomegalovirus, Epstein-Barr virus, or adenovirus. Primary endpoints were feasibility, safety, and clinical responses (>1 log reduction in viremia at 28 days). Secondary endpoints were reconstitution of antiviral immunity and persistence of the infused VSTs. Suitable VST products were identified for 75 of 77 clinical queries. Clinical responses were achieved in 29 of 47 (62%) of patients post-HSCT including 73% of patients evaluable at 1-month post-infusion, meeting the primary efficacy endpoint (>52%). Secondary graft rejection occurred in one child following VST infusion as described in a companion article. Corticosteroids, graft-versus-host disease, transplant-associated thrombotic microangiopathy, and eculizumab treatment correlated with poor response, while uptrending absolute lymphocyte and CD8 T cell counts correlated with good response. This study highlights key clinical factors that impact response to VSTs and demonstrates the feasibility and efficacy of this therapy in pediatric HSCT.

    View details for DOI 10.1038/s41467-024-47057-2

    View details for PubMedID 38637498

  • Radiation and Busulfan-Free Hematopoietic Stem Cell Transplantation Using Briquilimab (JSP191) Anti-CD117 Antibody-Conditioning, Transient Immunosuppression and TCR α β + T-Cell/CD19+B-Cell Depleted Haploidentical Grafts in Patients with Fanconi Anemia Agarwal, R., Bertaina, A., Soco, C., Saini, G., Kunte, N., Hiroshima, L., Chan, Y., Willner, H., Krampf, M. L., Nofal, R., Barbarito, G., Sen, S., Felber, M., Van Hentenryck, M., Walck, E., Scheck, A., Thongthip, S., Logan, A. C., Dougall, K., Bouge, A., Boelens, J., Long-Boyle, J. R., Weissman, I. L., Shizuru, J., Pang, W. W., Weinberg, K. I., Parkman, R., Roncarolo, M., Porteus, M., Czechowicz, A. AMER SOC HEMATOLOGY. 2023
  • LENTIVIRAL-MEDIATED GENE THERAPY FOR PATIENTS WITH FANCONI ANEMIA [GROUP A]: UPDATED RESULTS FROM GLOBAL RP-L102 CLINICAL TRIALS Sevilla, J., Booth, C., Czechowicz, A., Agarwal, R., Zubicaray, J., Rio, P., Chetty, K., O'Toole, G., Xu-Bayford, J., Ancliff, P., Sebastian, E., Choi, G., Zeini, M., Nicoletti, E., Eide, C., Wagner, J., Rao, G., Thrasher, A., Schwartz, J., Roncarolo, M., Bueren, J. SPRINGERNATURE. 2023: 276-277
  • Effect of Testicular Boost in Children With Leukemia Receiving Total Body Irradiation and Stem Cell Transplant: A Single-Institution Experience. Advances in radiation oncology Blomain, E. S., Jiang, A., Donaldson, S. S., Agarwal, R., Bertaina, A., Shyr, D., Eisenberg, M. L., Hoppe, R. T., Hiniker, S. M., Oh, J. 2023; 8 (1): 101071

    Abstract

    Children with leukemia who receive fractionated total body irradiation (fTBI) with 12 to 13.2 Gy as part of conditioning for hematopoietic stem cell transplant are frequently treated with an additional 4 Gy testicular boost to reduce the risk of testicular relapse. While institutional practices vary, limited data exists regarding whether the 4-Gy testicular boost reduces the risk of relapse and whether it causes toxicity beyond that imparted by TBI. This study compared the survival and endocrine outcomes among the patients who were treated with and without a testicular boost as part of fTBI from 1990 to 2019 at our center.We retrospectively reviewed charts of male children with leukemia treated with fTBI as part of a conditioning regimen for stem cell transplant from 1990 to 2019. Reported outcomes included progression-free survival, testicular relapse rate, and overall survival. Gonadal dysfunction and fertility were assessed by comparing the rate of abnormally low testosterone or high luteinizing hormone or follicular stimulating hormone, number of offspring, fertility service use, and abnormal sperm count in the subsequent follow-up period between the testicular boost and nonboost subset.Ninety-three male patients (63 acute lymphoblastic leukemia, 30 acute myeloid leukemia) with a median age of 9 years (range, 1-22) and follow-up of 3.3 years were included. In addition to 12- to 13.2-Gy fTBI, 51 male patients (54%) received a testicular boost to 4 Gy. There was 1 testicular relapse in the boost subset and none in the nonboost subset. Five-year progression-free survival for the boost and nonboost subset was 74% and 66%, respectively (P = .31). On multivariable analysis, boost was not associated with improved relapse-free survival or overall survival. More patients in the boost subset (35 of 51, 69%) had abnormal serum gonadal blood work compared with the nonboost subset (18 of 42, 43%) (P = .03).Omission of testicular boost may be associated with comparable oncologic but improved gonadal endocrine outcomes and should be further studied.

    View details for DOI 10.1016/j.adro.2022.101071

    View details for PubMedID 36483061

    View details for PubMedCentralID PMC9723295

  • LENTIVIRAL-MEDIATED GENE THERAPY FOR FANCONI ANEMIA [GROUP A]: RESULTS FROM RP-L102 CLINICAL TRIALS Czechowicz, A., Sevilla, J., Booth, C., Agarwal, R., Zubicaray, J., Rio, P., Navarro, S., Chetty, K., O'Toole, G., Xu-Bayford, J., Ancliff, P., Sebastian, E., Choi, G., Zeini, M., Nicoletti, E., Wagner, J., Eide, C., Rao, G., Thrasher, A., Schwartz, J., Roncarolo, M., Bueren, J. WILEY. 2023: S136-S137
  • Lentiviral-Mediated Gene Therapy for Fanconi Anemia [Group A]: Results from Global RP-L102 Clinical Trials Czechowicz, A., Sevilla, J., Booth, C., Navarro, S., Agarwal, R., Zubicaray, J., Rio, P., Chetty, K., O'Toole, G., Xu-Bayford, J., Ancliff, P., Sebastian, E., Choi, G., Zeini, M., Nicoletti, E., Eide, C., Wagner, J. E., Rao, G., Thrasher, A. J., Schwartz, J., Roncarolo, M., Bueren, J. A. CELL PRESS. 2023: 118
  • Effect of Testicular Boost in Children With Leukemia Receiving Total Body Irradiation and Stem Cell Transplant: A Single-Institution Experience ADVANCES IN RADIATION ONCOLOGY Blomain, E. S., Jiang, A., Donaldson, S. S., Agarwal, R., Bertaina, A., Shyr, D., Eisenberg, M. L., Hoppe, R. T., Hiniker, S. M., Oh, J. 2023; 8 (1)
  • Precision Delivery of Steroids as a Rescue Therapy for Gastrointestinal Graft-versus-Host Disease in Pediatric Stem Cell Transplant Recipients Journal of Clinical Medicine Levitte, s., Ganguly, A., Frolik, S., Guevara-Tique, A., et al 2023; 12 (4229)

    View details for DOI 10.3390/jcm12134229

  • Hematopoietic and Immunological Assessment in Fanconi Anemia after Ex Vivo Lentiviral FANCA Gene Therapy with RP-L102 Nofal, R., Chan, Y., Sen, S., Figueroa, U., Willner, H., Felber, M., Krampf, M., Thongthip, S., Choi, G., Nicoletti, E., Schwartz, J. D., Weinberg, K., Rodriguez, A., Agarwal, R., Roncarolo, M., Czechowicz, A. AMER SOC HEMATOLOGY. 2022: 7772-7773
  • Lentiviral-mediated Gene Therapy for Patients with Fanconi Anemia [Group A]: Updated Results from Global RP-L102 Clinical Trials Czechowicz, A., Sevilla, J., Booth, C., Agarwal, R., Zubicaray, J., Rio, P., Navarro, S., Chetty, K., O'Toole, G., Xu-Bayford, J., Ancliff, P., Sebastian, E., Choi, G., Zeini, M., Nicoletti, E., Wagner, J. E., Rao, G. R., Thrasher, A. J., Schwartz, J. D., Roncarolo, M., Bueren, J. A. AMER SOC HEMATOLOGY. 2022: 10646-10647
  • Sequential Stem Cell-Kidney Transplantation in Schimke Immuno-osseous Dysplasia. The New England journal of medicine Bertaina, A., Grimm, P. C., Weinberg, K., Parkman, R., Kristovich, K. M., Barbarito, G., Lippner, E., Dhamdhere, G., Ramachandran, V., Spatz, J. M., Fathallah-Shaykh, S., Atkinson, T. P., Al-Uzri, A., Aubert, G., van der Elst, K., Green, S. G., Agarwal, R., Slepicka, P. F., Shah, A. J., Roncarolo, M. G., Gallo, A., Concepcion, W., Lewis, D. B. 2022; 386 (24): 2295-2302

    Abstract

    Lifelong immunosuppression is required for allograft survival after kidney transplantation but may not ultimately prevent allograft loss resulting from chronic rejection. We developed an approach that attempts to abrogate immune rejection and the need for post-transplantation immunosuppression in three patients with Schimke immuno-osseous dysplasia who had both T-cell immunodeficiency and renal failure. Each patient received sequential transplants of alphabeta T-cell-depleted and CD19 B-cell-depleted haploidentical hematopoietic stem cells and a kidney from the same donor. Full donor hematopoietic chimerism and functional ex vivo T-cell tolerance was achieved, and the patients continued to have normal renal function without immunosuppression at 22 to 34 months after kidney transplantation. (Funded by the Kruzn for a Kure Foundation.).

    View details for DOI 10.1056/NEJMoa2117028

    View details for PubMedID 35704481

  • Sequential hematopoietic stem cell transplantation (HSCT) followed by kidney transplant-3 children who received ABT-cell/CD19 B-cell depleted HSCT followed by kidney transplant from same parental donor are rejection free and immunosuppression free with donor specific tolerance > 1year post kidney transplant Grimm, P., Lewis, D. B., Al-Uzri, A., Fathallah-Shaykh, S., Shah, A., Agarwal, R., Weinberg, K. I., Gallo, A., Concepcion, W., Bertaina, A. WILEY. 2022
  • Functional Immune Tolerance Induced By Sequential Hematopoietic Stem Cell-Solid Organ Transplantation Bertaina, A., Barbarito, G., Ramachandran, V. V., Kristovich, K., Lippner, E., Fathallah-Shaykh, S., Al-Uzri, A., Shah, A. J., Aubert, G., Slepicka, P., Oppizzi, L., Agarwal, R., Roncarolo, M., Gallo, A., Concepcion, W., Weinberg, K. I., Parkman, R., Lewis, D. B., Grimm, P. C. AMER SOC HEMATOLOGY. 2021: 1818-+
  • Gene Therapy for Fanconi Anemia [Group A]: Interim Results of RP-L102 Clinical Trials Czechowicz, A., Sevilla, J., Agarwal, R., Booth, C., Zubicaray, J., Rio, P., Navarro, S., Ancliff, P., Sebastian, E., Beard, B. C., Law, K. M., Choi, G., Zeini, M., Duran-Persson, C., Nicoletti, E., Wagner, J. E., Rao, G. R., Thrasher, A. J., Schwartz, J. D., Bueren, J. A., Roncarolo, M. AMER SOC HEMATOLOGY. 2021
  • Alloantigen-specific type 1 regulatory T cells suppress through CTLA-4 and PD-1 pathways and persist long-term in patients. Science translational medicine Chen, P. P., Cepika, A., Agarwal-Hashmi, R., Saini, G., Uyeda, M. J., Louis, D. M., Cieniewicz, B., Narula, M., Amaya Hernandez, L. C., Harre, N., Xu, L., Thomas, B. C., Ji, X., Shiraz, P., Tate, K. M., Margittai, D., Bhatia, N., Meyer, E., Bertaina, A., Davis, M. M., Bacchetta, R., Roncarolo, M. G. 2021; 13 (617): eabf5264

    Abstract

    [Figure: see text].

    View details for DOI 10.1126/scitranslmed.abf5264

    View details for PubMedID 34705520

  • Tissue-specific telomere shortening and degenerative changes in a patient with TINF2 mutation and dyskeratosis congenita. Human pathology (New York) Roake, C. M., Juntilla, M., Agarwal-Hashmi, R., Artandi, S., Kuo, C. S. 2021; 25

    Abstract

    Dyskeratosis congenita is a disease of impaired tissue maintenance downstream of telomere dysfunction. Characteristically, patients present with the clinical triad of nail dystrophy, oral leukoplakia, and skin pigmentation defects, but the disease involves degenerative changes in multiple organs. Mutations in telomere-binding proteins such as TINF2 (TRF1-interacting nuclear factor 2) or in telomerase, the enzyme that counteracts age related telomere shortening, are causative in dyskeratosis congenita. We present a patient who presented with severe hypoxemia at age 13. The patient had a history of myelodysplastic syndrome treated with bone marrow transplant at the age of 5. At age 18 she was hospitalized for an acute pneumonia progressing to respiratory failure, developed renal failure and ultimately, she and her family opted to withdraw support as she was not a candidate for a lung transplant. Sequencing of the patient's TINF2 locus revealed a heterozygous mutation (c.844C > T, Arg282Cys) which has previously been reported in a subset of dyskeratosis congenita patients. Tissue sections from multiple organs showed degenerative changes including disorganized bone remodeling, diffuse alveolar damage and small vessel proliferation in the lung, and hyperkeratosis with hyperpigmentation of the skin. Autopsy samples revealed a bimodal distribution of telomere length, with telomeres from donor hematopoietic tissues being an age-appropriate length and those from patient tissues showing pathogenic shortening, with the shortest telomeres in lung, liver, and kidney. We report for the first time a survey of degenerative changes and telomere lengths in multiple organs in a patient with dyskeratosis congenita.

    View details for DOI 10.1016/j.ehpc.2021.200517

    View details for PubMedID 34522616

  • Gene Therapy for Fanconi Anemia [Group A]: Preliminary Results of Ongoing RP-L102 Clinical Trials Czechowicz, A., Sevilla, J., Booth, C., Agarwal, R., Zubicaray, J., Rio, P., Navarro, S., Ancliff, P. J., Beard, B. C., Law, K. M., Choi, G., Zeini, M., Duran-Persson, C., Nicoletti, E., Rao, G. R., Wagner, J. E., Schwartz, J., Bueren, J. A., Roncarolo, M. CELL PRESS. 2021: 339
  • Preclinical Safety and Efficacy Validation of CD4(LVFOXP3) Cells as an Innovative Cell-Based Gene Therapy Approach for IPEX Syndrome Sato, Y., Nathan, A., Wright, J., Tate, K., Wani, P., Fazeli, F., Timnak, A., Bhatia, N., Agarwal-Hashmi, R., Bertaina, A., Roncarolo, M., Bacchetta, R. CELL PRESS. 2021: 340
  • Adoptively Transferred, In Vitro-Generated Alloantigen-Specific Type 1 Regulatory T (Tr1) Cells Persist Long-Term In Vivo Cepika, A., Chen, P. P., Agarwal, R., Saini, G., Louis, D. M., Amaya-Hernandez, L. C., Xu, L., Shiraz, P., Tate, K. M., Margittai, D., Bhatia, N., Meyer, E., Bertaina, A., Davis, M. M., Bacchetta, R., Roncarolo, M. CELL PRESS. 2021: 73
  • CD34 expression does not correlate with immunophenotypic stem cell or progenitor content in human cord blood products. Blood advances Mantri, S., Reinisch, A., Dejene, B. T., Lyell, D. J., DiGiusto, D. L., Agarwal-Hashmi, R., Majeti, R., Weinberg, K. I., Porteus, M. H. 2020; 4 (21): 5357–61

    View details for DOI 10.1182/bloodadvances.2020002891

    View details for PubMedID 33136125

  • The Binns Program for Cord Blood Research: A novel model of cord blood banking for academic biomedical research. Placenta Mantri, S., Sheikali, A., Binns, C., Lyell, D. J., DiGiusto, D. L., Porteus, M. H., Agarwal-Hashmi, R. 2020; 103: 50–52

    Abstract

    Umbilical cord blood is an important graft source in the treatment of many genetic, hematologic, and immunologic disorders by hematopoietic stem cell transplantation. Millions of cord blood units have been collected and stored for clinical use since the inception of cord blood banking in 1989. However, the use of cord blood in biomedical research has been limited by access to viable samples. Here, we present a cost-effective, self-sustaining model for the procurement of fresh umbilical cord blood components for research purposes within hospital-affiliated academic institutions.

    View details for DOI 10.1016/j.placenta.2020.10.018

    View details for PubMedID 33075720

  • A beta T-Cell/CD19 B-Cell Depleted Haploidentical Stem Cell Transplantation: A New Platform for Curing Rare and Monogenic Disorders Bertaina, A., Bacchetta, R., Lewis, D. B., Grimm, P. C., Shah, A. J., Agarwal, R., Concepcion, W., Czechowicz, A., Bhatia, N., Lahiri, P., Weinberg, K. I., Parkman, R., Porteus, M., Roncarolo, M. ELSEVIER SCIENCE INC. 2020: S288
  • The Binns Program for Cord Blood Research: A Novel Program for Cord Blood Procurement in an Academic Setting for Biomedical Research Sheikali, A., Mantri, S., Lyell, D., Porteus, M., Agarwal, R. ELSEVIER SCIENCE INC. 2020: S206
  • Regulatory Type 1 T Cell Infusion in Mismatched Related or Unrelated Hematopoietic Stem Cell Transplantation (HSCT) for Hematologic Malignancies Agarwal, R., Bacchetta, R., Bertaina, A., Chen, P., Saini, G., Shiraz, P., Bhatia, N., Roncarolo, M. ELSEVIER SCIENCE INC. 2020: S272–S273
  • Early Epigenetic Immune Quantification Following Alpha/Beta T-Cell/CD19 B-Cell Depleted Haploidentical Stem Cell Transplant Correlates with CD4+T Cell Recovery at Day+100 Mayers, M., Schulze, J., Barbarito, G., Lakshmanan, U., Parkman, R., Weinberg, K. I., Chu, J., Agarwal, R., Roncarolo, M., Sachsenmaier, C., Bacchetta, R., Bertaina, A. ELSEVIER SCIENCE INC. 2020: S305
  • Third-Party Virus-Specific T-Cell Infusion for Treatment of Refractory Viral Infections: Interim Results from PBMTC SUP1701 Keller, M. D., Hanley, P. J., Zhang, N., Tanna, J., Fatic, A., Lang, H., Ekanem, U., Sani, G. M., Aguayo-Hiraldo, P., Quigg, T. C., Verneris, M. R., Parikh, S., Dvorak, C. C., Satwani, P., Davila, B., Bednarski, J. J., Pai, S., Agarwal, R., Aquino, V., Smith, A. R., Gourdine, L., Bollard, C. M., Pulsipher, M. A. ELSEVIER SCIENCE INC. 2020: S89–S90
  • Alloantigen-specific Tr1 cells designed to prevent GvHD have a distinct molecular identity and suppress through CTLA-4 and PD-1 Society for Immunotherapy of Cancer’s (SITC) 35th Anniversary Annual Meeting Cepika, A., Chen, P. P., Uyeda, M. J., Cieniewicz, B., Narula, M., Amaya, L., Louis, D. M., Xu, L., Ji, X., Bertaina, A., Agarwal-Hashmi, R., Davis, M. M., Meyer, E., Bacchetta, R., Roncarolo, M. 2020: A159–A159
  • Brentuximab Vedotin as Consolidation Therapy After Autologous Stem Cell Transplantation in Children and Adolescents (<18y) With Early Relapse Hodgkin Lymphoma. Journal of pediatric hematology/oncology Fernandez, K. S., Mavers, M., Marks, L. J., Agarwal, R. 2019

    Abstract

    We describe 6 pediatric patients (12 to 18y) with relapsed or refractory Hodgkin lymphoma treated with consolidative Brentuximab vedotin (Bv) following reinduction chemotherapy and autologous stem cell transplantation. The progression-free survival after autologous stem cell transplantation was 12, 18, 22, 24, 30, and 30 months. Most patients tolerated Bv well although 2 patients developed grade 3 neuropathy that prevent them from completing the scheduled 16 doses of Bv. Consolidative Bv in children and adolescents, as currently recommended for adult patients with early relapsed or refractory Hodgkin lymphoma, is feasible but with some significant toxicities.

    View details for DOI 10.1097/MPH.0000000000001703

    View details for PubMedID 31876780

  • Non-Genotoxic Anti-CD117 Antibody Conditioning Results in Successful Hematopoietic Stem Cell Engraftment in Patients with Severe Combined Immunodeficiency Agarwal, R., Dvorak, C. C., Kwon, H., Long-Boyle, J. R., Prohaska, S. S., Brown, J. W., Le, A., Guttman-Klein, A., Weissman, I. L., Cowan, M. J., Logan, A. C., Weinberg, K. I., Parkman, R., Roncarolo, M., Shizuru, J. A. AMER SOC HEMATOLOGY. 2019
  • REGULATORY TYPE 1 T CELL INFUSION IN MISMATCHED RELATED OR UNRELATED HEMATOPOIETIC STEM CELL TRANSPLANTATION (HSCT) FOR HEMATOLOGIC MALIGNANCIES Agarwal, R., Bacchetta, R., Bertaina, A., Hu, J. C., Chen, P., Saini, G., Bhatia, N., Roncarolo, M. WILEY. 2019
  • Feasibility of brentuximab vedotin as consolidation therapy after autologous stem-cell transplantation in children and adolescents (< 18 years) with early relapse Hodgkin lymphoma. Fernandez, K. S., Mavers, M., Chang-Halpenny, C. N., Titapiwatanakun, R., Baker, K., Pugmire, B., Tcheng, W., Agarwal-Hashmi, R. AMER SOC CLINICAL ONCOLOGY. 2019
  • Chimerism Analysis in Pediatric Hematopoietic Stem Cell Transplantation for Non-Malignant Disorders Mariano, L., Zhang, B., Kristovich, K., Agarwal-Hashmi, R., Roncarolo, M., Bertaina, A., Fernandez-Vina, M. ELSEVIER SCIENCE INC. 2019
  • Toxicity-Free Hematopoietic Stem Cell Engraftment Achieved with Anti-CD117 Monoclonal Antibody Conditioning Agarwal, R., Dvorak, C. C., Prohaska, S., Long-Boyle, J., Kwon, H., Brown, J. M., Weinberg, K. I., Le, A., Guttman-Klein, A., Logan, A. C., Weissman, I. L., Digiusto, D., Cowan, M. J., Parkman, R., Roncarolo, M., Shizuru, J. A. ELSEVIER SCIENCE INC. 2019
  • Regulatory Type 1 T Cell Infusion in Mismatched Related or Unrelated Hematopoietic Stem Cell Transplantation (HSCT) for Hematologic Malignancies Agarwal, R., Bacchetta, R., Bertaina, A., Chu, J., Chen, P., Saini, G., Bhatia, N., Roncarolo, M. ELSEVIER SCIENCE INC. 2019
  • Administration of BPX-501 Cells Following A beta T and B-Cell-Depleted HLA Haploidentical HSCT (haplo-HSCT) in Children with Acute Leukemias (AL) Galaverna, F., Ruggeri, A., Merli, P., Kapoor, N., Agarwal-Hashmi, R., Aquino, V., Jacobsohn, D. A., Qasim, W., Nemecek, E. R., Krishnamurti, L., Manwani, D., Kuhn, M., Locatelli, F., Naik, S. ELSEVIER SCIENCE INC. 2019
  • Newborn Screening for Severe Combined Immunodeficiency and T-cell Lymphopenia in California, 2010-2017 PEDIATRICS Amatuni, G. S., Currier, R. J., Church, J. A., Bishop, T., Grimbacher, E., Nguyen, A., Agarwal-Hashmi, R., Aznar, C. P., Butte, M. J., Cowan, M. J., Dorsey, M. J., Dvorak, C. C., Kapoor, N., Kohn, D. B., Markert, M., Moore, T. B., Naides, S. J., Sciortino, S., Feuchtbaum, L., Koupaei, R. A., Puck, J. M. 2019; 143 (2)
  • Newborn Screening for Severe Combined Immunodeficiency and T-cell Lymphopenia in California, 2010-2017. Pediatrics Amatuni, G. S., Currier, R. J., Church, J. A., Bishop, T., Grimbacher, E., Nguyen, A. A., Agarwal-Hashmi, R., Aznar, C. P., Butte, M. J., Cowan, M. J., Dorsey, M. J., Dvorak, C. C., Kapoor, N., Kohn, D. B., Markert, M. L., Moore, T. B., Naides, S. J., Sciortino, S., Feuchtbaum, L., Koupaei, R. A., Puck, J. M. 2019

    Abstract

    OBJECTIVES: Newborn screening for severe combined immunodeficiency (SCID) was instituted in California in 2010. In the ensuing 6.5 years, 3252156 infants in the state had DNA from dried blood spots assayed for T-cell receptor excision circles (TRECs). Abnormal TREC results were followed-up with liquid blood testing for T-cell abnormalities. We report the performance of the SCID screening program and the outcomes of infants who were identified.METHODS: Data that were reviewed and analyzed included demographics, nursery summaries, TREC and lymphocyte flow-cytometry values, and available follow-up, including clinical and genetic diagnoses, treatments, and outcomes.RESULTS: Infants with clinically significant T-cell lymphopenia (TCL) were successfully identified at a rate of 1 in 15300 births. Of these, 50 cases of SCID, or 1 in 65000 births (95% confidence interval 1 in 51000-1 in 90000) were found. Prompt treatment led to 94% survival. Infants with non-SCID TCL were also identified, diagnosed and managed, including 4 with complete DiGeorge syndrome who received thymus transplants. Although no cases of typical SCID are known to have been missed, 2 infants with delayed-onset leaky SCID had normal neonatal TREC screens but came to clinical attention at 7 and 23 months of age.CONCLUSIONS: Population-based TREC testing, although unable to detect immune defects in which T cells are present at birth, is effective for identifying SCID and clinically important TCL with high sensitivity and specificity. The experience in California supports the rapid, widespread adoption of SCID newborn screening.

    View details for PubMedID 30683812

  • Central Nervous System Relapse After Stem Cell Transplantation in Adolescents and Young Adults with Acute Lymphoblastic Leukemia: A Single-Institution Experience. Journal of adolescent and young adult oncology Kozak, M. M., Yoo, C. H., Gutkin, P. M., von Eyben, R. n., Agarwal, R. n., Donaldson, S. S., Muffly, L. n., Hiniker, S. M. 2019

    Abstract

    Purpose: To evaluate outcomes and central nervous system (CNS) relapse in adolescents and young adults (AYAs) with acute lymphoblastic leukemia (ALL), who underwent total body irradiation (TBI) before allogeneic hematopoietic stem cell transplantation (allo-SCT). Methods: A total of 136 AYA patients with ALL who received TBI before allo-SCT between 1998 and 2018 were reviewed. Twenty patients received cranial radiation in their initial treatment before conditioning for transplant and were excluded. Competing risk analysis was used to estimate the cumulative incidence of relapse. Kaplan-Meier and log-rank tests were used to calculate overall survival (OS) and to identify factors predictive of relapse. OS and time to relapse were calculated from date of allo-SCT. Results: One hundred sixteen patients were included in the analysis. Median age was 27 years and median follow-up time was 42 months. Twenty-six patients suffered a disease relapse and 49 died, 26 of posttransplantation complications. The median time to relapse was 7 months and the 5-year OS was 60%. Seven patients had a CNS relapse: 4 of 20 patients (25%) with pre-SCT CNS disease had a post-allo-SCT CNS relapse compared to 3 of 97 (3.1%) without pre-SCT CNS disease. Median time to CNS relapse was 7 months. Patients with post-SCT CNS relapse had median OS of 19 months. Conclusions: AYA patients with CNS disease who undergo an allo-SCT have a high rate of CNS relapse. The addition of additional CNS-directed therapy to transplant protocols warrants further investigation.

    View details for DOI 10.1089/jayao.2019.0121

    View details for PubMedID 31747341

  • Related and unrelated donor transplantation for β-thalassemia major: results of an international survey. Blood advances Li, C. n., Mathews, V. n., Kim, S. n., George, B. n., Hebert, K. n., Jiang, H. n., Li, C. n., Zhu, Y. n., Keesler, D. A., Boelens, J. J., Dvorak, C. C., Agarwal, R. n., Auletta, J. J., Goyal, R. K., Hanna, R. n., Kasow, K. n., Shenoy, S. n., Smith, A. R., Walters, M. C., Eapen, M. n. 2019; 3 (17): 2562–70

    Abstract

    We studied 1110 patients with β-thalassemia major aged ≤25 years who received transplants with grafts from HLA-matched related (n = 677; 61%), HLA-mismatched related (n = 78; 7%), HLA-matched unrelated (n = 252; 23%), and HLA-mismatched unrelated (n = 103; 9%) donors between 2000 and 2016. Ninety percent of transplants were performed in the last decade. Eight-five percent of patients received ≥20 transfusions and 88% were inadequately chelated. All patients received myeloablative-conditioning regimen. Overall and event-free survival were highest for patients aged ≤6 years and after HLA-matched related and HLA-matched unrelated donor transplantation. The 5-year probabilities of overall survival for patients aged ≤6 years, 7 to 15 years, and 16 to 25 years, adjusted for donor type and conditioning regimen were 90%, 84%, and 63%, respectively (P < .001). The corresponding probabilities for event-free survival were 86%, 80%, and 63% (P < .001). Overall and event-free survival did not differ between HLA-matched related and HLA-matched unrelated donor transplantation (89% vs 87% and 86% vs 82%, respectively). Corresponding probabilities after mismatched related and mismatched unrelated donor transplantation were 73% vs 83% and 70% vs 78%. In conclusion, if transplantation is considered as a treatment option it should be offered early (age ≤6 years). An HLA-matched unrelated donor is a suitable alternative if an HLA-matched relative is not available.

    View details for DOI 10.1182/bloodadvances.2019000291

    View details for PubMedID 31471325

  • Administration of BPX-501 Cells Following A beta T and B-Cell-Depleted HLA Haploidentical HSCT (haplo-HSCT) in Children with Acute Leukemias Locatelli, F., Ruggeri, A., Merli, P., Naik, S., Agarwal, R., Aquino, V., Jacobsohn, D. A., Qasim, W., Nemecek, E. R., Krishnamurti, L., Manwani, D., Kuhn, M., Kapoor, N. AMER SOC HEMATOLOGY. 2018
  • Administration of Rimiducid Following Haploidentical BPX-501 Cells in Children with Malignant or Non-Malignant Disorders Who Develop Graft-Versus-Host-Disrase (GvHD) Elkeky, R., Jacobsohn, D. A., Agarwal, R., Naik, S., Kapoor, N., Krishnamurti, L., Slatter, M., Galaverna, F., Merli, P., Aldinger, M., Locatelli, F. AMER SOC HEMATOLOGY. 2018
  • Administration of BPX-501 Following alpha-T and B-Cell Depleted Haplo-HSCT in Children with Transfusion-Dependent Thalassemia Galaverna, F., Pagliara, D., Manwani, D., Agarwal, R., Kuhn, M., Locatelli, F. AMER SOC HEMATOLOGY. 2018
  • Related and Unrelated Donor Transplantation for beta Thalassemia Major: Results of an International Survey Li, C., Mathews, V., George, B., Kim, S., Hebert, K., Jiang, H., Li, C., Zhu, Y., Keesler, D. A., Agarwal, R., Boelens, J., Dvorak, C. C., Auletta, J., Goyal, R., Hanna, R., Kasow, K. A., Margolis, D., Shenoy, S., Walters, M. C., Eapen, M. AMER SOC HEMATOLOGY. 2018
  • Mapping the Cellular Heterogeneity of CD34-Selected Umbilical Cord Blood Products Mantri, S., Reinisch, A., Weinberg, K., Lyell, D., DiGiusto, D., Agarwal-Hashmi, R., Porteus, M. NATURE PUBLISHING GROUP. 2018: 505–7
  • The Binns Program for Cord Blood Research: a novel program for cord blood procurement in an academic setting for biomedical research Mantri, S., Lyell, D., DiGiusto, D., Porteus, M., Agarwal-Hashmi, R. NATURE PUBLISHING GROUP. 2018: 787–88
  • The Cost of Hematopoietic Stem-Cell Transplantation in the United States AMERICAN HEALTH AND DRUG BENEFITS Broder, M. S., Quock, T. P., Chang, E., Reddy, S. R., Agarwal-Hashmi, R., Arai, S., Villa, K. F. 2017; 10 (7): 366–73

    Abstract

    Hematopoietic stem-cell transplantation (HSCT) requires highly specialized, resource-intensive care. Myeloablative conditioning regimens used before HSCT generally require inpatient stays and are more intensive than other preparative regimens, and may therefore be more costly.To estimate the costs associated with inpatient HSCT according to the type of the conditioning regimen used and other potential contributors to the overall cost of the procedure.We used data from the Truven Health MarketScan insurance claims database to analyze healthcare costs for pediatric (age <18 years) and adult (age ≥18 years) patients who had autologous or allogeneic inpatient HSCT between January 1, 2010, and September 23, 2013. We developed an algorithm to determine whether conditioning regimens were myeloablative or nonmyeloablative/reduced intensity.We identified a sample of 1562 patients who had inpatient HSCT during the study period for whom the transplant type and the conditioning regimen were determinable: 398 patients had myeloablative allogeneic HSCT; 195 patients had nonmyeloablative/reduced-intensity allogeneic HSCT; and 969 patients had myeloablative autologous HSCT. The median total healthcare cost at 100 days was $289,283 for the myeloablative allogeneic regimen cohort compared with $253,467 for the nonmyeloablative/reduced-intensity allogeneic regimen cohort, and $140,792 for the myeloablative autologous regimen cohort. The mean hospital length of stay for the index (first claim of) HSCT was 35.6 days in the myeloablative allogeneic regimen cohort, 26.6 days in the nonmyeloablative/reduced-intensity allogeneic cohort, and 21.8 days in the myeloablative autologous regimen cohort.Allogeneic HSCT was more expensive than autologous HSCT, regardless of the regimen used. Myeloablative conditioning regimens led to higher overall costs than nonmyeloablative/reduced-intensity regimens in the allogeneic HSCT cohort, indicating a greater cost burden associated with inpatient services for higher-intensity preparative conditioning regimens. Pediatric patients had higher costs than adult patients. Future research should involve validating the algorithm for identifying conditioning regimens using clinical data.

    View details for PubMedID 29263771

  • Anti-Fungal Prophylaxis Using Intermediate Dose Ambisome is Associated with Delayed Methotrexate Clearance in Pediatric Patients Undergoing Allogeneic Hematopoietic Stem Cell Transplantation Schultz, L., Kumar, K., Stone, S., Callard, E., Witkowski, J., Shinn, L., Pinner, L., Weinberg, K. I., Porteus, M. H., Agarwal, R., Shah, A. J., Kharbanda, S. ELSEVIER SCIENCE INC. 2017: S297–S298
  • SINGLE INSTITUTION EXPERIENCE: COMPARISON OF HEPARIN VERSUS URSODIOL FOR PREVENTION OF VOD. Shinn, L., Pinner, L., Kula, J., Kumar, P., Agarwal, R., Weinberg, K. ONCOLOGY NURSING SOC. 2017
  • Anti-Fungal Prophylaxis Using Intermediate Dose Ambisome Is Associated with Delayed Methotrexate Clearance in Pediatric Patients Undergoing Allogeneic Hematopoietic Stem Cell Transplantation Schultz, L. M., Kumar, K., Stone, S., Callard, E., Witkowski, J., Shinn, L., Pinner, L., Franklin, S., Kula, J., Patel, N., Kumar, P., Weinberg, K. I., Porteus, M., Agarwal, R., Shah, A., Kharbanda, S. AMER SOC HEMATOLOGY. 2016
  • Invasive Fungal Disease in Pediatric Patients Undergoing Allogeneic Hematopoietic Stem Cell Transplant JOURNAL OF PEDIATRIC HEMATOLOGY ONCOLOGY Aftandilian, C., Weinberg, K., Willert, J., Kharbanda, S., Porteus, M., Maldonado, Y., Agarwal, R. 2016; 38 (7): 574-580
  • Invasive Fungal Disease in Pediatric Patients Undergoing Allogeneic Hematopoietic Stem Cell Transplant. Journal of pediatric hematology/oncology Aftandilian, C., Weinberg, K., Willert, J., Kharbanda, S., Porteus, M., Maldonado, Y., Agarwal, R. 2016; 38 (7): 574-580

    Abstract

    Invasive fungal disease (IFD) remains a major cause of morbidity and mortality in pediatric patients after allogeneic hematopoietic stem cell transplant (HSCT). We analyzed the outcome of 152 consecutive pediatric patients who underwent allogeneic HSCT from 2005 to 2012: 126 of these without a history of IFD and 26 with IFD before HSCT. Antifungal prophylaxis agent was determined by the primary transplant attending. The rate of IFD after HSCT among patients with or without prior IFD was similar (7.7% with and 7.1% without a history of fungal disease before transplant). Mortality in these 2 populations did not differ (35% vs. 28%, P=0.48, χ). Patients deemed at higher risk for IFD were generally placed on voriconazole prophylaxis; however, this did not affect rates of posttransplant IFD. All-cause mortality in patients with posttransplant IFD was significantly higher than those without posttransplant IFD (67% vs. 21%, P<0.0001,χ). Identifying risk factors for posttransplant IFD remains a high priority to improve outcome of HSCT.

    View details for DOI 10.1097/MPH.0000000000000629

    View details for PubMedID 27658021

  • Costs of Hematopoietic Stem Cell Transplantation and Associated Conditioning Regimens Quock, T. P., Broder, M. S., Chang, E., Reddy, S. R., Arai, S., Agarwal-Hashmi, R., Villa, K. F. ELSEVIER SCIENCE INC. 2016: S282
  • Costs of Hematopoietic Stem Cell Transplantation and Associated Conditioning Regimens Quock, T. P., Broder, M. S., Chang, E., Reddy, S. R., Arai, S., Agarwal-Hashmi, R., Villa, K. F. AMER SOC HEMATOLOGY. 2015
  • A Pediatric Case of T-Cell Prolymphocytic Leukemia PEDIATRIC BLOOD & CANCER Mitton, B., Coutre, S., Willert, J., Schlis, K., Porteus, M., Kharbanda, S., Agarwal-Hashmi, R. 2015; 62 (6): 1061-1062

    Abstract

    T-cell Prolymphocytic Leukemia (T-PLL) is a rare entity, and to date has never been reported in children. Here, we describe the first pediatric case of T-PLL in a 16-year old male and review his clinical course through treatment. He underwent therapy with alemtuzumab and pentostatin, which was successful in inducing initial remission. He then underwent an allogeneic matched sibling stem cell transplant following a myeloablative conditioning regimen and remains disease-free 1.5 years after diagnosis. Pediatr Blood Cancer © 2014 Wiley Periodicals, Inc.

    View details for DOI 10.1002/pbc.25336

    View details for PubMedID 25417638

  • A Reduced-Toxicity Regimen Is Associated with Durable Engraftment and Clinical Cure of Nonmalignant Genetic Diseases among Children Undergoing Blood and Marrow Transplantation with an HLA-Matched Related Donor. Biology of blood and marrow transplantation Mahadeo, K. M., Weinberg, K. I., Abdel-Azim, H., Miklos, D. B., Killen, R., Kohn, D., Crooks, G. M., Shah, A. J., Kharbanda, S., Agarwal, R., Kapoor, N. 2015; 21 (3): 440-444

    Abstract

    Blood and marrow transplantation (BMT) is a standard curative therapy for patients with nonmalignant genetic diseases. Myeloablative conditioning has been associated with significant regimen-related toxicity (RRT), whereas reduced-intensity conditioning regimens have been associated with graft failure. In this prospective pilot trial conducted at 2 centers between 2006 and 2013, we report the outcome of 22 patients with nonmalignant genetic diseases who were conditioned with a novel reduced-toxicity regimen: i.v. busulfan (16 mg/kg), alemtuzumab (52 mg/m(2)), fludarabine (140 mg/m(2)), and cyclophosphamide (105 mg/kg). The median age of the study population was 3.5 years (range, 5 months to 26 years). No cases of sinusoidal obstruction syndrome, severe or chronic graft-versus-host disease (GVHD), or primary graft failure were reported. Median time to neutrophil engraftment (>500 cells/μL) and platelet engraftment (>20K cells/μL) were 19 (range, 12 to 50) and 23.5 (range, 14 to 134) days, respectively. The median length of follow-up was 3 years (range, .2 to 6.3). The overall survival rates were 95% at 100 days (95% confidence interval, .72 to .99) and 90% at 6 years (95% confidence interval, .68 to .98). RRT and chronic GVHD are significant barriers to BMT for patients with nonmalignant genetic diseases. This alemtuzumab-based reduced-toxicity regimen appears to be promising with durable engraftment, effective cure of clinical disease, low rates of RRT, and no observed chronic GVHD.

    View details for DOI 10.1016/j.bbmt.2014.11.005

    View details for PubMedID 25459642

  • Improved outcomes after autologous bone marrow transplantation for children with relapsed or refractory hodgkin lymphoma: twenty years experience at a single institution. Biology of blood and marrow transplantation Garfin, P. M., Link, M. P., Donaldson, S. S., Advani, R. H., Luna-Fineman, S., Kharbanda, S., Porteus, M., Weinberg, K. I., Agarwal-Hashmi, R. 2015; 21 (2): 326-334

    Abstract

    The purpose of this study is to evaluate the survival of pediatric patients undergoing autologous bone marrow transplantation (auBMT) for relapsed or refractory Hodgkin lymphoma (rrHL) and to identify factors that might contribute to their outcome. We reviewed the records and clinical course of 89 consecutive rrHL patients ≤ 21 years old who underwent auBMT at Stanford Hospitals and Clinics and the Lucile Packard Children's Hospital, Stanford between 1989 and 2012. We investigated, by multiple analyses, patient, disease, and treatment characteristics associated with outcome. Endpoints were 5-year overall and event-free survival. Our findings include that cyclophosphamide, carmustine, and etoposide (CBV) as a conditioning regimen for auBMT is effective for most patients ≤ 21 years old with rrHL (5-year overall survival, 71%). Transplantation after the year 2001 was associated with significantly improved overall survival compared with our earlier experience (80% compared with 65%). Patients with multiply relapsed disease or with disease not responsive to initial therapy fared less well compared with those with response to initial therapy or after first relapse. Administration of post-auBMT consolidative radiotherapy (cRT) also appears to contribute to improved survival. We are able to conclude that high-dose chemotherapy with CBV followed by auBMT is effective for the treatment of rrHL in children and adolescents. Survival for patients who undergo auBMT for rrHL has improved significantly. This improvement may be because of patient selection and improvements in utilization of radiotherapy rather than improvements in chemotherapy. Further investigation is needed to describe the role of auBMT across the entire spectrum of patients with rrHL and to identify the most appropriate preparative regimen with or without cRT therapy in the treatment of rrHL in young patients.

    View details for DOI 10.1016/j.bbmt.2014.10.020

    View details for PubMedID 25445024

  • A Multidisciplinary Care Team Perspective on Children's Emotional Experience in Isolation for Stem Cell Transplantation 2015 BMT Tandem Meetings Savig, E. S., Gurevitch, J. H., Jackson, J. E., Malinowski, A., Ju, W. G., Leifer, L. J., Cohen, H. J., Sourkes, B. M., Agarwal-Hashmi, R. 2015: S180
  • Survival and neurocognitive outcomes after cranial or craniospinal irradiation plus total-body irradiation before stem cell transplantation in pediatric leukemia patients with central nervous system involvement. International journal of radiation oncology, biology, physics Hiniker, S. M., Agarwal, R., Modlin, L. A., Gray, C. C., Harris, J. P., Million, L., Kiamanesh, E. F., Donaldson, S. S. 2014; 89 (1): 67-74

    Abstract

    To evaluate survival and neurocognitive outcomes in pediatric acute lymphoblastic leukemia (ALL) patients with central nervous system (CNS) involvement treated according to an institutional protocol with stem cell transplantation (SCT) and a component of craniospinal irradiation (CSI) in addition to total-body irradiation (TBI) as preparative regimen.Forty-one pediatric ALL patients underwent SCT with TBI and received additional cranial irradiation or CSI because of CNS leukemic involvement. Prospective neurocognitive testing was performed before and after SCT in a subset of patients. Cox regression models were used to determine associations of patient and disease characteristics and treatment methods with outcomes.All patients received a cranial radiation boost; median total cranial dose was 24 Gy. Eighteen patients (44%) received a spinal boost; median total spinal dose for these patients was 18 Gy. Five-year disease-free survival (DFS) for all patients was 67%. Those receiving CSI had a trend toward superior DFS compared with those receiving a cranial boost alone (hazard ratio 3.23, P=.14). Patients with isolated CNS disease before SCT had a trend toward superior DFS (hazard ratio 3.64, P=.11, 5-year DFS 74%) compared with those with combined CNS and bone marrow disease (5-year DFS 59%). Neurocognitive testing revealed a mean post-SCT overall intelligence quotient of 103.7 at 4.4 years. Relative deficiencies in processing speed and/or working memory were noted in 6 of 16 tested patients (38%). Pre- and post-SCT neurocognitive testing revealed no significant change in intelligence quotient (mean increase +4.7 points). At a mean of 12.5 years after transplant, 11 of 13 long-term survivors (85%) had completed at least some coursework at a 2- or 4-year college.The addition of CSI to TBI before SCT in pediatric ALL with CNS involvement is effective and well-tolerated. Craniospinal irradiation plus TBI is worthy of further protocol investigation in children with CNS leukemia.

    View details for DOI 10.1016/j.ijrobp.2014.01.056

    View details for PubMedID 24725690

  • Unrelated donor allogeneic hematopoietic stem cell transplantation for patients with hemoglobinopathies using a reduced-intensity conditioning regimen and third-party mesenchymal stromal cells. Biology of blood and marrow transplantation Kharbanda, S., Smith, A. R., Hutchinson, S. K., McKenna, D. H., Ball, J. B., Lamb, L. S., Agarwal, R., Weinberg, K. I., Wagner, J. E. 2014; 20 (4): 581-586

    Abstract

    Allogeneic hematopoietic stem cell transplantation for patients with a hemoglobinopathy can be curative but is limited by donor availability. Although positive results are frequently observed in those with an HLA-matched sibling donor, use of unrelated donors has been complicated by poor engraftment, excessive regimen-related toxicity, and graft-versus-host disease (GVHD). As a potential strategy to address these obstacles, a pilot study was designed that incorporated both a reduced-intensity conditioning and mesenchymal stromal cells (MSCs). Six patients were enrolled, including 4 with high-risk sickle cell disease (SCD) and 2 with transfusion-dependent thalassemia major. Conditioning consisted of fludarabine (150 mg/m(2)), melphalan (140 mg/m(2)), and alemtuzumab (60 mg for patients weighing > 30 kg and .9 mg/kg for patients weighing <30 kg). Two patients received HLA 7/8 allele matched bone marrow and 4 received 4-5/6 HLA matched umbilical cord blood as the source of HSCs. MSCs were of bone marrow origin and derived from a parent in 1 patient and from an unrelated third-party donor in the remaining 5 patients. GVHD prophylaxis consisted of cyclosporine A and mycophenolate mofetil. One patient had neutropenic graft failure, 2 had autologous hematopoietic recovery, and 3 had hematopoietic recovery with complete chimerism. The 2 SCD patients with autologous hematopoietic recovery are alive. The remaining 4 died either from opportunistic infection, GVHD, or intracranial hemorrhage. Although no infusion-related toxicity was seen, the cotransplantation of MSCs was not sufficient for reliable engraftment in patients with advanced hemoglobinopathy. Although poor engraftment has been observed in nearly all such trials to date in this patient population, there was no evidence to suggest that MSCs had any positive impact on engraftment. Because of the lack of improved engraftment and unacceptably high transplant-related mortality, the study was prematurely terminated. Further investigations into understanding the mechanisms of graft resistance and development of strategies to overcome this barrier are needed to move this field forward.

    View details for DOI 10.1016/j.bbmt.2013.12.564

    View details for PubMedID 24370862

  • Cytokine and chemokine patterns across 100 days after hematopoietic stem cell transplantation in children. Biology of blood and marrow transplantation DiCarlo, J., Agarwal-Hashmi, R., Shah, A., Kim, P., Craveiro, L., Killen, R., Rosenberg-Hasson, Y., Maecker, H. 2014; 20 (3): 361-369

    Abstract

    We mapped the cytokine response to hematopoietic stem cell transplantation (HSCT) by assaying 51 cytokines and chemokines each week for 100 days in 51 children receiving allogeneic (n = 44) or autologous HSCT (n = 7). Assay values were reported as mean fluorescence intensity (MFI). Log transformation converted MFI to clinically relevant measures (ie, pg/mL). We searched for potential markers of transplant complications by using mixed treatment by subject analysis of variance. Global cytokine secretion in HSCT recipients was significantly lower than in concurrent control patients (n = 11). Coincident with the nadir in WBC count, the concentration of many cytokines declined further by the second and third week. All analytes (except monokine induced by gamma interferon [MIG]) subsequently rebounded by week 4 (coincident with engraftment and recovery of WBC count) but often still remained well below control levels. Concurrent with the collective nadir of multiple cytokines, monocyte chemoattractant protein 1 (MCP-1), growth-regulated oncogene alpha (GRO-a), and leptin surged during weeks 2 to 4. High levels of leptin persisted throughout the 100 post-transplant days. Also during weeks 2 to 4, hepatocyte growth factor (HGF) and IL-6 surged in children with complications but not in those without complications. The peak in HGF was more pronounced in veno-occlusive disease (VOD). HGF and IL-6 secretion rose at least 2 weeks before the clinical diagnosis of VOD or graft-versus-host disease (GVHD). From week 4 onward in all groups, the MFI of the cytokine resistin increased to 5 to 15 times above concurrent control. HGF has now emerged in 3 or more biomarker discovery efforts for GVHD (and in our population for VOD as well). HGF (with or without IL-6) should be investigated as a potential predictive biomarker of VOD or GVHD. Alternatively, the hyperinflammatory "signature" provided by a multicytokine assay may be predictive.

    View details for DOI 10.1016/j.bbmt.2013.11.026

    View details for PubMedID 24316459

  • Outcome in Pediatric Patients with a History of Fungal Disease Prior to Allogeneic Stem Cell Transplant Aftandilian, C., Agarwal, R., Kharbanda, S., Weinberg, K. I. ELSEVIER SCIENCE INC. 2014: S165
  • END Organ Disease in the Context of Human Herpes VIRUS 6 Viremia in Pediatric Allogeneic Hematopoietic STEM CELL Transplant Patients: A Case Series Winestone, L., Agarwal, R., Montoya, J., Weinberg, K. I., Porteus, M. H., Pinsky, B., Soda, E., Waggoner, J., Tamaresis, J., Kharbanda, S. ELSEVIER SCIENCE INC. 2014: S255
  • Survival and Neurocognitive Outcomes Following Cranial or Craniospinal Irradiation Plus Total Body Irradiation Prior to Transplantation in Children with CNS Leukemia Hiniker, S. M., Agarwal, R., Modlin, L. A., Harris, J. P., Kiamanesh, E. E., Million, L., Gray, C. C., Donaldson, S. S. ELSEVIER SCIENCE INC. 2014: S170–S171
  • Incidence and Mortality of Invasive Fungal Disease in Pediatric Patients after Allogeneic Stem Cell Transplant Aftandilian, C., Agarwal, R., Kharbanda, S., Weinberg, K. I. ELSEVIER SCIENCE INC. 2014: S165
  • Durable Engraftment, Correction of Genetic Defects and Prevention of Veno-Occlussive Disease, Following Blood and Marrow Transplantation with an HLA-Matched Sibling DONOR, Using a Reduced Toxicity Conditioning Regimen with Busulfan, Reduced Dose Cyclophos Mahadeo, K., Agarwal, R., Weinberg, K. I., Abdel-Azim, H., Miklos, D. B., Shah, A. J., Tabba, L., Kapoor, N. ELSEVIER SCIENCE INC. 2014: S83
  • Newborn screening for severe combined immunodeficiency and T-cell lymphopenia in California: Results of the first 2 years JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY Kwan, A., Church, J. A., Cowan, M. J., Agarwal, R., Kapoor, N., Kohn, D. B., Lewis, D. B., McGhee, S. A., Moore, T. B., Stiehm, E. R., Porteus, M., Aznar, C. P., Currier, R., Lorey, F., Puck, J. M. 2013; 132 (1): 140-U245

    Abstract

    Assay of T-cell receptor excision circles (TRECs) in dried blood spots obtained at birth permits population-based newborn screening (NBS) for severe combined immunodeficiency (SCID).We sought to report the first 2 years of TREC NBS in California.Since August 2010, California has conducted SCID NBS. A high-throughput TREC quantitative PCR assay with DNA isolated from routine dried blood spots was developed. Samples with initial low TREC numbers had repeat DNA isolation with quantitative PCR for TRECs and a genomic control, and immunophenotyping was performed within the screening program for infants with incomplete or abnormal results. Outcomes were tracked.Of 993,724 infants screened, 50 (1/19,900 [0.005%]) had significant T-cell lymphopenia. Fifteen (1/66,250) required hematopoietic cell or thymus transplantation or gene therapy; these infants had typical SCID (n = 11), leaky SCID or Omenn syndrome (n = 3), or complete DiGeorge syndrome (n = 1). Survival to date in this group is 93%. Other T-cell lymphopenic infants had variant SCID or combined immunodeficiency (n = 6), genetic syndromes associated with T-cell impairment (n = 12), secondary T-cell lymphopenia (n = 9), or preterm birth (n = 8). All T-cell lymphopenic infants avoided live vaccines and received appropriate interventions to prevent infections. TREC test specificity was excellent: only 0.08% of infants required a second test, and 0.016% required lymphocyte phenotyping by using flow cytometry.TREC NBS in California has achieved early diagnosis of SCID and other conditions with T-cell lymphopenia, facilitating management and optimizing outcomes. Furthermore, NBS has revealed the incidence, causes, and follow-up of T-cell lymphopenia in a large diverse population.

    View details for DOI 10.1016/j.jaci.2013.04.024

    View details for Web of Science ID 000321052300019

    View details for PubMedID 23810098

  • Bortezomib As a Treatment for Autoimmune Cytopenia After Bone Marrow Transplant BMT Tandem Meetings Lalefar, N. R., Agarwal, R., Porteus, M. H., Kharbanda, S., Weinberg, K. I. ELSEVIER SCIENCE INC. 2013: S177–S177
  • Autologous Transplantation for Hodgkin disease: A Tale of Two Eras BMT Tandem Meetings Garfin, P. M., Luna-Fineman, S., Amylon, M., Kharbanda, S., Weinberg, K. I., Willert, J. R., Porteus, M., Link, M., Agarwal, R. ELSEVIER SCIENCE INC. 2013: S247–S247
  • Single Institution Experience With Allogeneic Hematopoietic Stem Cell Transplantation for Pediatric and AYA Patients With Acute Leukemia BMT Tandem Meetings Breese, E., Agarwal, R., Kharbanda, S., Breese, M., Amylon, M., Weinberg, K. I., Porteus, M., Dahl, G., Lacayo, N., Schlis, K., Willert, J. R. ELSEVIER SCIENCE INC. 2013: S242–S243
  • Hematopoietic Cell Transplantation (HCT) for Treatment of Genetic Lymphohematopoietic Diseases for Patients Lacking a Fully Matched Sibling Donor Using a Novel Conditioning Regimen BMT Tandem Meetings Kharbanda, S., Agarwal, R., Miklos, D. B., Porteus, M., Amylon, M., Willert, J. R., Weinberg, K. I. ELSEVIER SCIENCE INC. 2013: S250–S251
  • High Risk Allogeneic Hematopoietic Cell Transplant (HCT) Patients with Any Level of Cytomegalovirus (CMV) Viremia Should Be Treated with Antiviral Therapy to Prevent Serious CMV Disease BMT Tandem Meetings Winestone, L., Agarwal, R., Weinberg, K. L., Porteus, M., Willerts, J. R., Amylon, M., Kharbanda, S. ELSEVIER SCIENCE INC. 2013: S312–S312
  • ENDOCRINE SEQUELAE OF STEM CELL TRANSPLANTATION Barnum, J., Agarwal, R., Bachrach, L. WILEY PERIODICALS, INC. 2012: 1022–23
  • THE CYTOKINE RESPONSE TO HEMATOPOIETIC STEM CELL TRANSPLANTATION 40th Critical Care Congress Kim, P., Agarwal, R., Shah, A., Craver, L., Killen, R., Rosenberg-Hasson, Y., DiCarlo, J. LIPPINCOTT WILLIAMS & WILKINS. 2010: U80–U80
  • Pathological evidence of Wolman's disease following hematopoietic stem cell transplantation despite correction of lysosomal acid lipase activity BONE MARROW TRANSPLANTATION Gramatges, M. M., Dvorak, C. C., Regula, D. P., Enns, G. M., Weinberg, K., Agarwal, R. 2009; 44 (7): 449-450

    View details for DOI 10.1038/bmt.2009.57

    View details for Web of Science ID 000270933000007

    View details for PubMedID 19308038

  • Nontuberculous Mycobacteria Infections in Immunocompromised Patients Single Institution Experience JOURNAL OF PEDIATRIC HEMATOLOGY ONCOLOGY Wei, M. C., Banaei, N., Yakrus, M. A., Stoll, T., Gutierrez, K. M., Agarwal, R. 2009; 31 (8): 556-560

    Abstract

    Disseminated infection due to nontuberculous Mycobacterium (NTM) species is rare in pediatrics. Here we report 6 infections affecting 5 patients at a single institution in an immunocompromised population of pediatric oncology and stem cell transplant recipients. The patients presented within a 1-year period with catheter-associated bacteremia. New pulmonary nodules were noted in 4 of the 5 patients. All of the infections were due to rapidly growing NTM. Patients were successfully treated with removal of the infected catheter and combination antibiotic therapy. There are currently no consensus guidelines for treatment of NTM infections in this population, and a therapeutic approach is presented here.

    View details for PubMedID 19641470

  • High-dose chemotherapy followed by stem cell rescue for high-risk germ cell tumors: the Stanford experience BONE MARROW TRANSPLANTATION Agarwal, R., Dvorak, C. C., Stockerl-Goldstein, K. E., Johnston, L., Srinivas, S. 2009; 43 (7): 547-552

    Abstract

    Germ cell tumors carry an excellent prognosis with platinum-based therapy upfront. The patients who either relapse or demonstrate refractoriness to platinum pose a challenge. There exist many reports in the literature on the use of high-dose chemotherapy and stem cell rescue improving the outcome in patients with relapsed germ cell tumors. However, the reports have great variability in the patient selection, prior treatments, the choice of the conditioning regimen and variability of the doses within the same regimen. In this report, we present 37 patients who underwent a uniform protocol of high-dose chemotherapy with stem cell rescue. Stem cell mobilization was performed with high-dose CY (4 g per m(2)) and we were able to collect adequate cells for marrow rescue in all patients. Patients received a high-dose regimen with etoposide (800 mg/m(2) per day) days -6, -5 and -4 as a continuous infusion, carboplatin (667 mg/m(2) per day) on days -6, -5 and -4 as a 1 h infusion, and CY (60 mg/kg per day) on days -3 and -2. In this high-risk group of patients, high-dose chemotherapy with autologous stem cell rescue led to a 3-year overall survival of 57% and a 3-year event-free survival of 49%. The results are reflective of a single procedure. No tandem transplants were performed. The treatment-related mortality was low at 3% in this heavily pretreated group.

    View details for DOI 10.1038/bmt.2008.364

    View details for Web of Science ID 000265005800005

    View details for PubMedID 18997833

  • Delayed Platelet Engraftment and Early Increased Creatinine after Stem Cell Transplant Predicts Sustained Remission in Pediatric Leukemia Sridhar, M., Dvorak, C. C., Agarwal, R., Schiffman, J. D. AMER SOC HEMATOLOGY. 2008: 752
  • Hematopoietic stem cell transplant for pediatric acute promyelocytic leukemia BIOLOGY OF BLOOD AND MARROW TRANSPLANTATION Dvorak, C. C., Agarwal, R., Dahl, G. V., Gregory, J. J., Feusner, J. H. 2008; 14 (7): 824-830

    Abstract

    The optimal form of treatment for children with relapsed or refractory acute promyelocytic leukemia (APL) is unclear. We retrospectively analyzed the results of 32 (11 autologous, 21 allogeneic) hematopoietic stem cell transplants (HSCT) performed for children originally treated on either the Eastern Cooperative Group E2491 Trial or the Cancer and Leukemia Group B C9710 Trial and subsequently diagnosed with relapsed or refractory APL. For autologous HSCT, the incidence of treatment-related mortality (TRM) and relapse was 0% (95% confidence interval [CI], 0%-30%) and 27% (95% CI, 9%-57%), respectively. The 5-year event-free survival (EFS) and overall survival (OS) following autologous HSCT was 73% (95% CI, 43%-91%) and 82% (95% CI, 51%-96%), respectively. For allogeneic HSCT, the incidence of TRM and relapse was 19% (95% CI, 7%-41%) and 10% (95% CI, 2%-30%), respectively. The 5-year EFS and OS following allogeneic HSCT was 71% (95% CI, 50%-86%) and 76% (95% CI, 55%-90%), respectively. There was no significant difference in EFS or OS between autologous and allogeneic HSCT. This data demonstrates that autologous and allogeneic HSCT are both effective therapies for treatment of children with relapsed or refractory APL. Autologous HSCT is associated with a low incidence of TRM, whereas allogeneic HSCT is associated with a low incidence of relapse, suggesting a strong GVL effect against residual APL.

    View details for DOI 10.1016/j.bbmt.2008.04.015

    View details for Web of Science ID 000256971000013

    View details for PubMedID 18541203

    View details for PubMedCentralID PMC2796449

  • SAFETY OF HEMATOPOIETIC STEM CELL TRANSPLANTATION IN CHILDREN LESS THAN THREE YEARS OF AGE PEDIATRIC HEMATOLOGY AND ONCOLOGY Dvorak, C. C., Wright, N. B., Wong, W. B., Kristovich, K. M., Matthews, E. W., Weinberg, K. I., Amylon, M. D., Agarwal, R. 2008; 25 (8): 705-722

    Abstract

    Allogeneic hematopoietic stem cell transplantation (HSCT) is a standard treatment for a variety of hematologic conditions. However, very young children may experience different complications of HSCT compared to older patients. The authors retrospectively analyzed the results of 51 transplants performed on children less than 3 years of age between June 1987 and October 2005. Donors were matched-related (n = 21), partially mismatched related (n = 3), and unrelated (n = 27). The majority of patients had one or more grade III organ toxicities, but all nonrelapse deaths were attributable to infection. Perineal dermatitis was found in a large number (73%) of recipients of cyclophosphamide-based conditioning regimens. The 1-year transplant-related mortality (TRM) was 14%, but significantly declined in the more modern period. Grades II-IV acute graft-versus-host-disease (GvHD) was seen in 22% of patients, while chronic extensive GvHD developed in only 7% of patients. Relapse was seen in 40% of transplants performed for a malignant condition, most commonly in those patients not in remission at time of HSCT. The 5-year event-free survival (EFS) and overall survival (OS) were 53 and 64%, respectively. Recipients of fractionated total body irradiation (fTBI) were more likely to have at least one long-term sequelae than patients who received chemotherapy-based regimens (p = .014). These data demonstrate that HSCT can be performed safely in very young children, especially as supportive-care techniques improve. Cyclophosphamide-related perineal dermatitis is a unique complication in very young children. Finally, the incidence of acute and chronic GvHD in this population is low.

    View details for DOI 10.1080/08880010802243524

    View details for PubMedID 19065437

  • Optimization of conditioning for marrow transplantation from unrelated donors for patients with aplastic anemia after failure of immunosuppressive therapy BLOOD Deeg, H. J., O'Donnell, M., Tolar, J., Agarwal, R., Harris, R. E., Feig, S. A., Territo, M. C., Collins, R. H., McSweeney, P. A., Copelan, E. A., Khan, S. P., Woolfrey, A., Storer, B. 2006; 108 (5): 1485-1491

    Abstract

    In 87 patients with aplastic anemia who failed to respond to immunosuppressive treatment, we determined the minimal dose of total body irradiation (TBI) required when added to antithymocyte globulin (ATG, 30 mg/kg x 3) plus cyclophosphamide (CY, 50 mg/kg x 4) to achieve engraftment of unrelated donor marrow. TBI was started at 3 x 200 cGy, to be escalated or deescalated in steps of 200 cGy depending on graft failure or toxicity. Patients were aged 1.3 to 53.5 years (median, 18.6 years). The interval from diagnosis to transplantation was 3 to 328 months (median, 14.6 months). Donors were HLA-A, -B, -C, -DR, and -DQ identical for 62 patients, and nonidentical for 1 to 3 HLA loci at the antigen or allele level for 25. The dose-limiting toxicity was diffuse pulmonary injury. The optimum TBI dose was 1 x 200 cGy. Nine patients did not tolerate ATG and were prepared with CY + TBI. Graft failure occurred in 5% of patients. With a median follow-up of 7 years, 38 (61%) of 62 HLA-identical, and 10 (40%) of 25 HLA-nonidentical transplant recipients are surviving. The highest survival rate with HLA-identical transplants was observed at 200 cGy TBI. Thus, low-dose TBI + CY + ATG conditioning resulted in excellent outcome of unrelated transplants in patients with aplastic anemia who had received multiple transfusions.

    View details for DOI 10.1182/blood-2006-03-005041

    View details for Web of Science ID 000240271500016

    View details for PubMedID 16684959

    View details for PubMedCentralID PMC1895515

  • Use of intravenous mycophenolate mofetil for graft-versus-host disease prophylaxis in an allogeneic hematopoietic stem cell transplant recipient with an allergic reaction to cyclosporine and tacrolimus BONE MARROW TRANSPLANTATION Dvorak, C. C., Callard, E., Agarwal, R. 2006; 38 (3): 253-254

    View details for DOI 10.1038/sj.bmt.1705422

    View details for Web of Science ID 000239118600014

    View details for PubMedID 16785867

  • Risks and outcomes of invasive fungal infections in pediatric patients undergoing allogeneic hematopoietic cell transplantation BONE MARROW TRANSPLANTATION Dvorak, C. C., Steinbach, W. J., Brown, J. M., Agarwal, R. 2005; 36 (7): 621-629

    Abstract

    Invasive fungal infections (IFI) are the leading cause of infectious mortality in adult patients undergoing hematopoietic cell transplantation (HCT) after myeloablative conditioning, but the extent of this problem in the pediatric population is unclear. We retrospectively examined risk factors for IFI among 120 consecutive pediatric patients undergoing allogeneic HCT at a single center. The incidence of proven or probable IFI in pediatric patients during the first year after allogeneic HCT was 13%, comparable to the rate reported in adult patients; however, unlike IFI in adult patients, the majority of IFI in children occurred within the first month after transplantation. The primary risk factors for IFI were duration of neutropenia, age greater than 10 years, transplant for severe aplastic anemia or Fanconi anemia, and high-dose corticosteroid administration for 10 days or longer. IFI were more likely to be successfully treated (42%, 5/12 patients) in pediatric HCT recipients when compared to previous reports of adult recipients. Nonrelapse mortality was estimated at 17% (20/120 patients) after allogeneic HCT, of which 35% (seven patients) were directly attributed to IFI. Thus, IFI is a significant cause of nonrelapse mortality in children undergoing allogeneic HCT and more effective strategies are needed to prevent and treat IFI.

    View details for DOI 10.1038/sj.bmt.1705113

    View details for Web of Science ID 000231877200009

    View details for PubMedID 16044133

  • High-dose therapy and autologous hematopoietic stem-cell transplantation for recurrent or refractory pediatric Hodgkin's disease: Results and prognostic indices 45th Annual Meeting of the American-Society-for-Therapeutic-Radiology-and-Oncology (ASTRO) Lieskovsky, Y. E., Donaldson, S. S., Torres, M. A., Wong, R. M., Amylon, M. D., Link, M. P., Agarwal, R. AMER SOC CLINICAL ONCOLOGY. 2004: 4532–40

    Abstract

    To evaluate the outcome of pediatric patients with refractory or relapsed Hodgkin's disease (HD) who undergo high-dose therapy and autologous hematopoietic stem-cell transplantation (AHSCT).From 1989 to 2001, 41 pediatric patients with relapsed or primary refractory HD underwent high-dose therapy followed by AHSCT according to one of four autologous transplantation protocols at Stanford University Medical Center (Stanford, CA). Pretreatment factors were analyzed by univariate and multivariate analysis for prognostic significance for 5-year overall survival (OS), event-free survival (EFS), and progression-free survival (PFS).At a median follow-up of 4.2 years (range, 0.7 to 11.9 years), the 5-year OS, EFS, and PFS rates were 68%, 53%, and 63%, respectively. Multivariate analysis determined the following three factors to be significant predictors of poor OS and EFS: extranodal disease at first relapse, presence of mediastinal mass at time of AHSCT, and primary induction failure. Two of these factors also predicted for poor PFS (extranodal disease at time of first relapse and presence of mediastinal mass at time of transplantation).More than half of children with relapsed or refractory HD can be successfully treated with the combination of high-dose therapy and AHSCT, confirming the efficacy of this approach. Further investigation is now required to determine the optimal timing of AHSCT, as well as to develop alternative regimens for those patients with factors prognostic for poor outcome after AHSCT.

    View details for DOI 10.1200/JCO.2004.02.121

    View details for PubMedID 15542804

  • Continuous veno-venous hemofiltration may improve survival from acute respiratory distress syndrome after bone marrow transplantation or chemotherapy JOURNAL OF PEDIATRIC HEMATOLOGY ONCOLOGY DiCarlo, J. V., Alexander, S. R., Agarwal, R., Schiffman, J. D. 2003; 25 (10): 801-805

    Abstract

    Acute respiratory distress syndrome (ARDS) may result from immunologic activity triggered by irradiation and/or chemotherapy. Hemofiltration removes plasma water and soluble components below 25 kilodaltons. The authors hypothesized that early hemofiltration might attenuate the inflammatory component of ARDS, resulting in increased survival in immunocompromised children and young adults.Ten children (6 bone marrow transplantation, 3 chemotherapy, 1 lymphoma/hemophagocytosis) with ARDS (Pao2/Fio2 94 +/- 37 torr) received early continuous veno-venous hemodiafiltration as adjunctive therapy for respiratory failure, regardless of renal function. Six children had normal urine output and initial serum creatinine (range 0.1-1.2 mg/dL); four had renal insufficiency (initial creatinine 1.7-2.4 mg/dL). Hemofiltration was instituted coincident with intubation. Respiratory failure was precipitated by Enterobacter sepsis in two patients and by Aspergillus in one.Hemodiafiltration was performed for 13 +/- 9 days. A high rate of clearance was achieved (52 +/- 17 mL/min/1.73 m2). Duration of mechanical ventilation was 14 +/- 9 days. Nine of the 10 children were successfully extubated; 8 survived.Early hemofiltration may improve survival from ARDS following bone marrow transplantation or chemotherapy. Possible mechanisms include strict fluid balance, immunomodulation through filtration of inflammatory constituents, and immunomodulation through intensive extracellular water exchange that delivers biochemicals to organs of metabolism as well as the hemofilter.

    View details for PubMedID 14528104

  • Early CVVH increases survival from bone marrow transplant ARDS DiCarlo, J. V., Alexander, Agarwal-Hashmi, R., Schiffman, J. D. INT PEDIATRIC RESEARCH FOUNDATION, INC. 2003: 274A–275A
  • Antithrombin-III for the treatment of chemotherapy-induced organ dysfunction following bone marrow transplantation BONE MARROW TRANSPLANTATION Morris, J. D., Harris, R. E., Hashmi, R., SAMBRANO, J. E., Gruppo, R. A., Becker, A. T., Morris, C. L. 1997; 20 (10): 871-878

    Abstract

    A hypercoaguable state has been shown to follow high-dose chemotherapy for bone marrow transplantation (BMT). Deficiency of the natural anticoagulants, antithrombin III (AT-III), protein C and protein S correlate with organ dysfunction following BMT. We treated 10 patients with severe post-BMT organ dysfunction with AT-III concentrate. Indications for treatment included AT-III anticoagulant level less than 88% and life-threatening single or multiorgan dysfunction. All patients were loaded with 50 units/kg AT-III every 8 h for three doses followed by 50 units/kg/day each day for 3-12 days. Clinical improvement was seen within 1-5 days of start of therapy in all patients. Patients with veno-occlusive disease (VOD) showed a decrease in platelet consumption in nine of nine patients, resolution of hepatic tenderness in six of eight patients, and reduction of severe ascites and weight gain in four of five patients. The probability of death due to VOD and life-threatening organ dysfunction was significantly less in the AT-III-treated group when compared to a historical control group receiving the same preparative regimen (P = 0.047 and P = 0.034, respectively). Significant improvements in organ dysfunction following AT-III treatment in this small study supports a causal relationship between AT-III deficiency and post-BMT chemotherapy-induced organ dysfunction.

    View details for Web of Science ID A1997YG89600010

    View details for PubMedID 9404929

  • Retroviral transduction of CD34-enriched hematopoietic progenitor cells under serum-free conditions HUMAN GENE THERAPY Sekhar, M., Kotani, H., Doren, S., Agarwal, R., McGarrity, G., DUNBAR, C. E. 1996; 7 (1): 33-38

    Abstract

    The use of defined or serum-free culture conditions during retroviral transduction of hematopoietic cells would be desirable for standardization and safety reasons, as well as potentially allowing greater expansion of progenitor cells. Retroviral vector supernatants were concentrated and purified via tangential flow filtration polyethylene glycol (PEG)-precipitation, and ultracentrifugation, allowing serum-free transductions at standard multiplicities of infection (moi). Protein content of transductions using these concentrated vectors was 5-6 logs lower than in standard transductions. Transduction efficiencies of these concentrated vector preparations added back to serum-free or serum-containing media were equivalent to standard retroviral supernatant transductions of CD34-enriched progenitors. Absolute progenitor (CFU-C) numbers at the end of transduction were higher in serum-free + concentrated virus transductions, as opposed to transductions in standard vector supernatants containing fetal calf serum.

    View details for Web of Science ID A1996UA84500004

    View details for PubMedID 8825866

  • LONG-TERM CULTURE OF CHRONIC MYELOGENOUS LEUKEMIA MARROW-CELLS ON STEM-CELL FACTOR-DEFICIENT STROMA FAVORS BENIGN PROGENITORS BLOOD Agarwal, R., Doren, S., Hicks, B., DUNBAR, C. E. 1995; 85 (5): 1306-1312

    Abstract

    Long-term culture of marrow from patients with chronic myelogenous leukemia (CML) has been reported to favor the outgrowth of bcr/abl- progenitor cells in some patients. We examined the effect of the presence of soluble or transmembrane forms of stem cell factor (SCF) in long-term cultures of CML marrow. CD34-enriched cells from CML patients in advanced chronic phase or accelerated phase were plated on immortalized fetal liver stromal cells from homozygous SCF-deficient SI/SI mice (SI/SI4) with or without the addition of soluble human SCF, SI/SI4 cells expressing high levels of the transmembrane form of human SCF (SI/SIh220), or primary human allogeneic stroma. Cells were removed from cultures and plated weekly in colony assays. The clonagenic cell output from cultures completely lacking SCF was lower over the first 2 to 3 weeks, but by 5 weeks was similar to the clonagenic cell output from the other culture conditions. Analysis of bcr/abl transcripts from individual colonies showed a lower percentage of malignant progenitors present in long-term cultures completely deficient in SCF than under the other culture conditions, particularly compared with primary human stroma-containing long-term cultures. SCF may specifically favor malignant versus benign progenitor cells present in the marrow of CML patients, and an abnormal proliferative response to SCF in very primitive cells may be an underlying defect in the pathophysiology of this disease.

    View details for Web of Science ID A1995QJ43500021

    View details for PubMedID 7532038

  • SPLENIC AND HEPATIC PELIOSIS - MR FINDINGS AMERICAN JOURNAL OF ROENTGENOLOGY MAVES, C. K., CARON, K. H., Bisset, G. S., Agarwal, R. 1992; 158 (1): 75-76

    View details for Web of Science ID A1992GW60800014

    View details for PubMedID 1727362