88 results on '"Forman, S."'
Search Results
2. Reduced-Intensity Allogeneic Peripheral Blood Stem Cell Transplantation for High-Risk Acute Lymphoblastic Leukemia: A Potential Therapeutic Approach for High Risk ALL Patients Not Eligible for Full Intensity Transplantation
- Author
-
Stein, A., primary, Palmer, J, primary, O’Donnell, M, primary, Kogut, N., primary, Spielberger, R, primary, Slovak, ML, primary, Tsai, NC, primary, and Forman, S. J, primary
- Published
- 2008
- Full Text
- View/download PDF
3. Clinical Outcomes of Patients with T Cell NHL Undergoing Allogeneic Stem Cell Transplant.
- Author
-
Zain, Jasmine, primary, Palmer, J., additional, Tsai, N., additional, Popplewell, L., additional, Nadamanee, A., additional, Krishnan, A., additional, Nakamura, R., additional, Rodriguez, R., additional, Forman, S. J., additional, and Kirschbaum, M., additional
- Published
- 2006
- Full Text
- View/download PDF
4. Allogeneic Hematopoietic Cell Transplantation (HCT) with Nonmyeloablative Conditioning after Failed Myeloablative HCT: Factors Affecting Outcomes.
- Author
-
Baron, Frederic, primary, Storb, R., primary, Storer, B., primary, Maloney, D., primary, Maris, M., primary, Niederwieser, D., primary, Shizuru, J., primary, Chauncey, T., primary, Bruno, B., primary, Forman, S., primary, McSweeney, P., primary, Maziarz, R., primary, Pulsipher, M., primary, Agura, E., primary, Sorror, M., primary, Wade, J., primary, and Sandmaier, B., primary
- Published
- 2005
- Full Text
- View/download PDF
5. Efficacy of Nonmyeloablative Hematopoietic Cell Transplant (HCT) in Secondary Myelodysplastic Syndrome (MDS) and Its Impact on the Primary Disease.
- Author
-
Kerbauy, F., primary, Maris, M., primary, Storer, B., primary, Maloney, D., primary, Niederwieser, D., primary, Agura, E., primary, Pulsipher, M., primary, Chauncey, T., primary, Maziarz, R., primary, Forman, S., primary, Langston, A., primary, Wade, J., primary, Scott, B., primary, Deeg, J., primary, Storb, R., primary, and Sandmaier, B. M., primary
- Published
- 2005
- Full Text
- View/download PDF
6. HLA-Matched Related (MRD) or Unrelated Donor (URD) Non-Myeloablative Hematopoietic Cell Transplantation (HCT) for Patients (pts) with Refractory and Relapsed Aggressive Non Hodgkin Lymphoma (NHL).
- Author
-
Norasetthada, L., primary, Maris, M. B., primary, Sandmaier, B. M., primary, Gooley, T., primary, Forman, S., primary, Agura, E., primary, Maziarz, R. T., primary, Shizuru, J., primary, Niederwieser, D., primary, Storb, R., primary, and Maloney, D. G., primary
- Published
- 2004
- Full Text
- View/download PDF
7. Analysis of Long Term Outcome in Autologous Stem Cell Transplant (ASCT) for Acute Myelogenous Leukemia (AML) in First Remission (CR1) - a 15 Year Experience.
- Author
-
Stein, Anthony Selwyn, primary, O’Donnell, M., additional, Parker, P., additional, Spielberger, R., additional, Nademanee, A., additional, Bhatia, R., additional, Snyder, D. S., additional, Kirschbaum, M., additional, Pullarkat, V., additional, Kogut, N., additional, Bolotin, E., additional, Chang, K., additional, Sarkodee-Adoo, C., additional, Slovak, M., additional, Vora, N., additional, Land, J., additional, Dagis, A., additional, Palmer, J., additional, and Forman, S. J., additional
- Published
- 2004
- Full Text
- View/download PDF
8. Peripheral Blood Stem Cells vs Bone Marrow for Matched Sibling Transplant in AML and ALL in First Remission.
- Author
-
Kirschbaum, Mark, primary, O’Donnell, M., additional, Spielberger, R., additional, Bhatia, R., additional, Pullarkat, V., additional, Stein, A. S., additional, Kogut, N., additional, Bolotin, E., additional, Chang, K., additional, Land, J., additional, Sarkodee-Adoo, C., additional, Snyder, D., additional, and Forman, S. J., additional
- Published
- 2004
- Full Text
- View/download PDF
9. Outcomes for Unrelated Donor Hematopoietic Cell Transplantation (URD-HCT) for Patients with Acute Lymphoblastic Leukemia (ALL) Using Fractionated Total Body Irradiation (FTBI) and Etoposide (VP16).
- Author
-
O'Donnell, Margaret, Parker, P, Dagis, A, Slovak, ML, Snyder, D, Stein, A, Spielberger, R., Pawlowska, A, Rosenthal, J, Palmer, J, Kogut, N., and Forman, S. J
- Published
- 2008
- Full Text
- View/download PDF
10. Time course of reversibility of accelerated fibrinogen disappearance in diabetes mellitus: association with intravascular volume shifts
- Author
-
Jones, RL, Jovanovic, L, Forman, S, and Peterson, CM
- Abstract
Accelerated fibrinogen disappearance in diabetic patients is reversible with normalization of blood glucose. To define the time course of this reversal, we measured 125I-fibrinogen disappearance in 19 diabetic patients experiencing acute changes in blood glucose, as monitored and controlled by a microprocessor-controlled closed loop insulin infusion system (artificial beta cell). The data were corrected for blood volume dilutional changes and fit to a model describing two sequential exponential functions and a single exponential function. The sequential model provided the best fit for all but one patient. This indicates that there were two distinct rates of fibrinogen disappearance and suggests that the time course of reversal of accelerated fibrinogen disappearance in diabetic patients is very rapid, if not immediate. Rapid fibrinogen turnover during hyperglycemia was temporally associated with vascular volume changes, reflected as dilutional changes of 51Cr-RBC concentrations. These findings were also associated with an increase in pulse pressure during hyperglycemia, suggesting blood volume expansion due to an osmotic mechanism. The results of this study suggest a picture of vascular volume expansion and contraction, perhaps secondary to the osmotic effects of hyperglycemia. Accelerated fibrinogen turnover associated with these events may be related to increased vascular permeability and/or increased fibrin formation. These events, in concert, may contribute to the initiation and/or propagation of diabetic vascular sequelae.
- Published
- 1984
- Full Text
- View/download PDF
11. Pembrolizumab plus vorinostat induces responses in patients with Hodgkin lymphoma refractory to prior PD-1 blockade.
- Author
-
Mei M, Chen L, Godfrey J, Song J, Egelston C, Puverel S, Budde LE, Armenian S, Nikolaenko L, Nwangwu M, Guo W, Gao L, Lee P, Chen R, Daniels S, Kennedy N, Peters L, Zain J, Rosen S, Forman S, Popplewell L, Kwak L, and Herrera AF
- Subjects
- Adult, Humans, Vorinostat, Programmed Cell Death 1 Receptor therapeutic use, Neoplasm Recurrence, Local, Hodgkin Disease drug therapy, Antibodies, Monoclonal, Humanized
- Abstract
This phase 1 study evaluated the addition of vorinostat to pembrolizumab in patients with relapsed/refractory (RR) classical Hodgkin lymphoma (cHL), diffuse large B-cell lymphoma, and follicular lymphoma. We report the results in cases of cHL. Adult patients with RR cHL who had received ≥1 prior lines of therapy and were ineligible for transplantation were treated in a dose-escalation cohort with 2 dose levels (DLs) and then on an expansion cohort at the recommended phase 2 dose (RP2D) in 21-day cycles. Vorinostat 100 mg twice a day (DL1) and 200 mg twice a day (DL2) was administered orally from days 1 to 5 and 8 to 12; all patients received pembrolizumab 200 mg IV every 3 weeks. The primary end point was safety and determination of RP2D. In total, 32 patients with cHL were enrolled, including 30 at DL2 (RP2D); 78% had received prior anti-programmed cell death 1 (anti-PD-1) therapy, and 56% were PD-1 refractory. Grade ≥3 adverse events (AEs) included hypertension (9%), neutropenia (9%), hypophosphatemia (9%), thrombocytopenia (6%), and lymphopenia (6%). Immune-related AEs included grade 1 or 2 thyroiditis (13%), grade 1 rash (6%), and grade 3 esophagitis/duodenitis (3%). The overall response rate (ORR) was 72% and complete response (CR) rate was 34%. Patients refractory to prior PD-1 blockade (n = 18) had ORR and CR rates of 56% and 11%, respectively. Pembrolizumab and vorinostat was well tolerated with a high ORR rate in RR cHL including in anti-PD-1-refractory disease. This trial was registered at www.clinicaltrials.gov as #NCT03150329., (© 2023 by The American Society of Hematology.)
- Published
- 2023
- Full Text
- View/download PDF
12. Antibodies from donor B cells perpetuate cutaneous chronic graft-versus-host disease in mice.
- Author
-
Jin H, Ni X, Deng R, Song Q, Young J, Cassady K, Zhang M, Forman S, Martin PJ, Liu Q, and Zeng D
- Subjects
- Animals, B-Lymphocyte Subsets metabolism, B-Lymphocyte Subsets transplantation, Chemokine CCL20 metabolism, Chronic Disease, Dendritic Cells metabolism, Graft vs Host Disease pathology, Immunoglobulin G analysis, Immunoglobulin Heavy Chains, Immunoglobulin gamma-Chains genetics, Immunoglobulin gamma-Chains immunology, Immunoglobulin mu-Chains genetics, Immunoglobulin mu-Chains immunology, Interleukin-23 metabolism, Lymphoid Tissue pathology, Mice, Mice, Inbred BALB C, Mice, Inbred DBA, Radiation Chimera, Skin pathology, Specific Pathogen-Free Organisms, Th17 Cells immunology, Thymus Gland pathology, B-Lymphocyte Subsets immunology, Graft vs Host Disease immunology, Isoantibodies immunology
- Abstract
Cutaneous sclerosis is one of the most common clinical manifestations of chronic graft-versus-host disease (cGVHD). Donor CD4(+) T and B cells play important roles in cGVHD pathogenesis, but the role of antibodies from donor B cells remains unclear. In the current studies, we generated immunoglobulin (Ig)H(µγ1) DBA/2 mice whose B cells have normal antigen-presentation and regulatory functions but cannot secrete antibodies. With a murine cGVHD model using DBA/2 donors and BALB/c recipients, we have shown that wild-type (WT) grafts induce persistent cGVHD with damage in the thymus, peripheral lymphoid organs, and skin, as well as cutaneous T helper 17 cell (Th17) infiltration. In contrast, IgH(µγ1) grafts induced only transient cGVHD with little damage in the thymus or peripheral lymph organs or with little cutaneous Th17 infiltration. Injections of IgG-containing sera from cGVHD recipients given WT grafts but not IgG-deficient sera from recipients given IgH(µγ1) grafts led to deposition of IgG in the thymus and skin, with resulting damage in the thymus and peripheral lymph organs, cutaneous Th17 infiltration, and perpetuation of cGVHD in recipients given IgH(µγ1) grafts. These results indicate that donor B-cell antibodies augment cutaneous cGVHD in part by damaging the thymus and increasing tissue infiltration of pathogenic Th17 cells., (© 2016 by The American Society of Hematology.)
- Published
- 2016
- Full Text
- View/download PDF
13. Serum-resistant CpG-STAT3 decoy for targeting survival and immune checkpoint signaling in acute myeloid leukemia.
- Author
-
Zhang Q, Hossain DM, Duttagupta P, Moreira D, Zhao X, Won H, Buettner R, Nechaev S, Majka M, Zhang B, Cai Q, Swiderski P, Kuo YH, Forman S, Marcucci G, and Kortylewski M
- Subjects
- Animals, Cell Line, Tumor, Cell Survival drug effects, Cell Survival immunology, Drug Stability, Genes, cdc immunology, Genetic Therapy methods, Humans, Mice, Mice, Inbred C57BL, Mice, Transgenic, Molecular Targeted Therapy, Oligodeoxyribonucleotides chemistry, Oligodeoxyribonucleotides therapeutic use, STAT3 Transcription Factor chemistry, STAT3 Transcription Factor genetics, Serum physiology, Signal Transduction drug effects, CpG Islands, Genes, cdc drug effects, Leukemia, Myeloid, Acute immunology, Leukemia, Myeloid, Acute pathology, Leukemia, Myeloid, Acute therapy, Oligodeoxyribonucleotides pharmacology, STAT3 Transcription Factor antagonists & inhibitors, Tumor Escape drug effects
- Abstract
Targeting oncogenic transcription factor signal transducer and activator of transcription 3 (STAT3) in acute myeloid leukemia (AML) can reduce blast survival and tumor immune evasion. Decoy oligodeoxynucleotides (dODNs), which comprise STAT3-specific DNA sequences are competitive inhibition of STAT3 transcriptional activity. To deliver STAT3dODN specifically to myeloid cells, we linked STAT3dODN to the Toll-like receptor 9 (TLR9) ligand, cytosine guanine dinucleotide (CpG). The CpG-STAT3dODN conjugates are quickly internalized by human and mouse TLR9(+)immune cells (dendritic cells, B cells) and the majority of patients' derived AML blasts, including leukemia stem/progenitor cells. Following uptake, CpG-STAT3dODNs are released from endosomes, and bind and sequester cytoplasmic STAT3, thereby inhibiting downstream gene expression in target cells. STAT3 inhibition in patients' AML cells limits their immunosuppressive potential by reduced arginase expression, thereby partly restoring T-cell proliferation. Partly chemically modified CpG-STAT3dODNs have >60 hours serum half-life which allows for IV administration to leukemia-bearing mice (50% effective dose ∼ 2.5 mg/kg). Repeated administration of CpG-STAT3dODN resulted in regression of human MV4-11 AML in mice. The antitumor efficacy of this strategy is further enhanced in immunocompetent mice by combining direct leukemia-specific cytotoxicity with immunogenic effects of STAT3 blocking/TLR9 triggering. CpG-STAT3dODN effectively reducedCbfb/MYH11/MplAML burden in various organs and eliminated leukemia stem/progenitor cells, mainly through CD8/CD4 T-cell-mediated immune responses. In contrast, small-molecule Janus kinase 2/STAT3 inhibitor failed to reproduce therapeutic effects of cell-selective CpG-STAT3dODN strategy. These results demonstrate therapeutic potential of CpG-STAT3dODN inhibitors with broad implications for treatment of AML and potentially other hematologic malignancies., (© 2016 by The American Society of Hematology.)
- Published
- 2016
- Full Text
- View/download PDF
14. Role of altered growth factor receptor-mediated JAK2 signaling in growth and maintenance of human acute myeloid leukemia stem cells.
- Author
-
Cook AM, Li L, Ho Y, Lin A, Li L, Stein A, Forman S, Perrotti D, Jove R, and Bhatia R
- Subjects
- Animals, Antigens, CD34 metabolism, Cell Proliferation drug effects, Cell Survival drug effects, Cell Survival genetics, Disease Models, Animal, Female, Gene Expression Regulation, Leukemic, Humans, Janus Kinase 2 antagonists & inhibitors, Janus Kinases metabolism, Leukemia, Myeloid, Acute genetics, Leukemia, Myeloid, Acute pathology, Mice, Neoplastic Stem Cells drug effects, Phenotype, Phosphorylation, Pyrazoles pharmacology, Pyrimidines pharmacology, RNA Interference, Receptors, Growth Factor genetics, STAT Transcription Factors metabolism, STAT3 Transcription Factor genetics, STAT3 Transcription Factor metabolism, STAT5 Transcription Factor genetics, STAT5 Transcription Factor metabolism, Xenograft Model Antitumor Assays, Janus Kinase 2 metabolism, Leukemia, Myeloid, Acute metabolism, Neoplastic Stem Cells metabolism, Receptors, Growth Factor metabolism, Signal Transduction drug effects
- Abstract
Acute myeloid leukemia (AML) is sustained by small populations of leukemia stem cells (LSCs) that can resist available treatments and represent important barriers to cure. Although previous studies have shown increased signal transducer and activator of transcription (STAT)3 and STAT5 phosphorylation in AML leukemic blasts, the role of Janus kinase (JAK) signaling in primary AML compared with normal stem cells has not been directly evaluated. We show here that JAK/STAT signaling is increased in LSCs, particularly from high-risk AML. JAK2 inhibition using small molecule inhibitors or interference RNA reduced growth of AML LSCs while sparing normal stem cells both in vitro and in vivo. Increased JAK/STAT activity was associated with increased expression and altered signaling through growth factor receptors in AML LSCs, including receptor tyrosine kinase c-KIT and FMS-related tyrosine kinase 3 (FLT3). Inhibition of c-KIT and FLT3 expression significantly inhibited JAK/STAT signaling in AML LSCs, and JAK inhibitors effectively inhibited FLT3-mutated AML LSCs. Our results indicate that JAK/STAT signaling represents an important signaling mechanism supporting AML LSC growth and survival. These studies support continued evaluation of strategies for JAK/STAT inhibition for therapeutic targeting of AML LSCs.
- Published
- 2014
- Full Text
- View/download PDF
15. Leukemia cell-targeted STAT3 silencing and TLR9 triggering generate systemic antitumor immunity.
- Author
-
Hossain DM, Dos Santos C, Zhang Q, Kozlowska A, Liu H, Gao C, Moreira D, Swiderski P, Jozwiak A, Kline J, Forman S, Bhatia R, Kuo YH, and Kortylewski M
- Subjects
- Animals, CD8-Positive T-Lymphocytes cytology, Cell Line, Tumor, CpG Islands, Gene Silencing, Immune Tolerance, Interferon-gamma metabolism, Mice, Mice, Inbred C57BL, Mice, Inbred NOD, Mice, Knockout, Mice, SCID, Mice, Transgenic, RNA, Small Interfering metabolism, STAT3 Transcription Factor metabolism, Gene Expression Regulation, Leukemic, Leukemia immunology, Leukemia metabolism, STAT3 Transcription Factor genetics, Toll-Like Receptor 9 metabolism
- Abstract
Signal transducer and activator of transcription 3 (STAT3) is an oncogene and immune checkpoint commonly activated in cancer cells and in tumor-associated immune cells. We previously developed an immunostimulatory strategy based on targeted Stat3 silencing in Toll-like receptor 9 (TLR9)-positive hematopoietic cells using CpG-small interfering RNA (siRNA) conjugates. Here, we assessed the therapeutic effect of systemic STAT3 blocking/TLR9 triggering in disseminated acute myeloid leukemia (AML). We used mouse Cbfb-MYH11/Mpl-induced leukemia model, which mimics human inv(16) AML. Our results demonstrate that intravenously delivered CpG-Stat3 siRNA, but not control oligonucleotides, can eradicate established AML and impair leukemia-initiating potential. These antitumor effects require host's effector T cells but not TLR9-positive antigen-presenting cells. Instead, CpG-Stat3 siRNA has direct immunogenic effect on AML cells in vivo upregulating major histocompatibility complex class-II, costimulatory and proinflammatory mediators, such as interleukin-12, while downregulating coinhibitory PD-L1 molecule. Systemic injections of CpG-Stat3 siRNA generate potent tumor antigen-specific immune responses, increase the ratio of tumor-infiltrating CD8(+) T cells to regulatory T cells in various organs, and result in CD8(+) T-cell-dependent regression of leukemia. Our findings underscore the potential of using targeted STAT3 inhibition/TLR9 triggering to break tumor tolerance and induce immunity against AML and potentially other TLR9-positive blood cancers.
- Published
- 2014
- Full Text
- View/download PDF
16. TLR9-mediated siRNA delivery for targeting of normal and malignant human hematopoietic cells in vivo.
- Author
-
Zhang Q, Hossain DM, Nechaev S, Kozlowska A, Zhang W, Liu Y, Kowolik CM, Swiderski P, Rossi JJ, Forman S, Pal S, Bhatia R, Raubitschek A, Yu H, and Kortylewski M
- Subjects
- Animals, Cell Line, Tumor, Gene Transfer Techniques, Hematologic Neoplasms genetics, Hematopoietic Stem Cells pathology, Humans, Leukemia, Myeloid, Acute genetics, Leukemia, Myeloid, Acute therapy, Lymphoma genetics, Lymphoma therapy, Mice, Mice, Inbred BALB C, Mice, Inbred NOD, Mice, Knockout, Mice, SCID, Multiple Myeloma genetics, Multiple Myeloma therapy, Neoplasm Transplantation, Oligodeoxyribonucleotides genetics, RNA, Small Interfering genetics, Radiotherapy, STAT3 Transcription Factor genetics, Toll-Like Receptor 9 metabolism, Transplantation, Heterologous, Genetic Therapy methods, Hematologic Neoplasms therapy, Hematopoietic Stem Cells physiology, RNA, Small Interfering pharmacokinetics, Toll-Like Receptor 9 genetics
- Abstract
STAT3 operates in both cancer cells and tumor-associated immune cells to promote cancer progression. As a transcription factor, it is a highly desirable but difficult target for pharmacologic inhibition. We have recently shown that the TLR9 agonists CpG oligonucleotides can be used for targeted siRNA delivery to mouse immune cells. In the present study, we demonstrate that a similar strategy allows for targeted gene silencing in both normal and malignant human TLR9(+) hematopoietic cells in vivo. We have developed new human cell-specific CpG(A)-STAT3 siRNA conjugates capable of inducing TLR9-dependent gene silencing and activation of primary immune cells such as myeloid dendritic cells, plasmacytoid dendritic cells, and B cells in vitro. TLR9 is also expressed by several human hematologic malignancies, including B-cell lymphoma, multiple myeloma, and acute myeloid leukemia. We further demonstrate that oncogenic proteins such as STAT3 or BCL-X(L) are effectively knocked down by specific CpG(A)-siRNAs in TLR9(+) hematologic tumor cells in vivo. Targeting survival signaling using CpG(A)-siRNAs inhibits the growth of several xenotransplanted multiple myeloma and acute myeloid leukemia tumors. CpG(A)-STAT3 siRNA is immunostimulatory and nontoxic for normal human leukocytes in vitro. The results of the present study show the potential of using tumoricidal/immunostimulatory CpG-siRNA oligonucleotides as a novel 2-pronged therapeutic strategy for hematologic malignancies.
- Published
- 2013
- Full Text
- View/download PDF
17. S1PR1 is an effective target to block STAT3 signaling in activated B cell-like diffuse large B-cell lymphoma.
- Author
-
Liu Y, Deng J, Wang L, Lee H, Armstrong B, Scuto A, Kowolik C, Weiss LM, Forman S, and Yu H
- Subjects
- Animals, Apoptosis drug effects, B-Lymphocytes drug effects, B-Lymphocytes pathology, Cell Line, Tumor, Cell Proliferation drug effects, Cell Survival drug effects, Disease Models, Animal, Fingolimod Hydrochloride, Gene Silencing drug effects, Humans, Lymphocyte Activation drug effects, Lymphoma, Large B-Cell, Diffuse immunology, Lymphoma, Large B-Cell, Diffuse pathology, Mice, Neoplasm Invasiveness, Phosphorylation drug effects, Propylene Glycols pharmacology, RNA, Small Interfering metabolism, Receptors, Lysosphingolipid antagonists & inhibitors, Signal Transduction drug effects, Sphingosine analogs & derivatives, Sphingosine pharmacology, Sphingosine-1-Phosphate Receptors, B-Lymphocytes immunology, Lymphocyte Activation immunology, Lymphoma, Large B-Cell, Diffuse metabolism, Receptors, Lysosphingolipid metabolism, STAT3 Transcription Factor metabolism
- Abstract
STAT3 plays a crucial role in promoting progression of human cancers, including several types of B-cell lymphoma. However, as a transcription factor lacking its own enzymatic activity, STAT3 remains difficult to target with small-molecule drugs in the clinic. Here we demonstrate that persistent activated STAT3 colocalizes with elevated expression of S1PR1, a G-protein-coupled receptor for sphingosine-1-phosphate (S1P), in the tumor cells of the activated B cell-like subtype of diffuse large B-cell lymphoma patient specimens. Inhibition of S1PR1 expression by shRNA in the lymphoma cells validates that blocking S1PR1 affects expression of STAT3 downstream genes critically involved in tumor cell survival, proliferation, tumor invasion, and/or immunosuppression. Using S1PR1 shRNA, or FTY720, an antagonist of S1P that is in the clinic for other indications, we show that inhibiting S1PR1 expression down-regulates STAT3 activity and causes growth inhibition of the lymphoma tumor cells in vitro and in vivo. Our results suggest that targeting S1P/S1PR1 using a clinically relevant and available drug or other approaches is potentially an effective new therapeutic modality for treating the activated B cell-like subtype of diffuse large B-cell lymphoma, a subset of lymphoma that is less responsive to current available therapies.
- Published
- 2012
- Full Text
- View/download PDF
18. Phase 1/2 trial of total marrow and lymph node irradiation to augment reduced-intensity transplantation for advanced hematologic malignancies.
- Author
-
Rosenthal J, Wong J, Stein A, Qian D, Hitt D, Naeem H, Dagis A, Thomas SH, and Forman S
- Subjects
- Adolescent, Adult, Aged, Bone Marrow drug effects, Bone Marrow pathology, Child, Feasibility Studies, Female, Graft vs Host Disease etiology, Hematologic Neoplasms pathology, Humans, Lymph Nodes drug effects, Lymph Nodes pathology, Male, Melphalan administration & dosage, Middle Aged, Pilot Projects, Prospective Studies, Survival Rate, Transplantation Conditioning, Transplantation, Homologous, Treatment Outcome, Vidarabine administration & dosage, Vidarabine analogs & derivatives, Young Adult, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Bone Marrow radiation effects, Graft vs Host Disease prevention & control, Hematologic Neoplasms therapy, Lymph Nodes radiation effects, Peripheral Blood Stem Cell Transplantation
- Abstract
This phase 1/2 study assessed the augmentation of reduced-intensity conditioning (RIC) with total marrow and lymph node irradiation (TMLI), for peripheral blood stem cell transplantation, in patients with advanced hematologic disease. The regimen consisted of fludarabine 25 mg/m(2) per day for 5 days, melphalan 140 mg/m(2) for one day, and TMLI radiation at 150 cGy/fraction in 8 fractions over 4 days. Eligible patients were over 50 years old and/or had compromised organ function. Median age of the 33 evaluable patients was 55.2 years. Eighteen events of nonhematologic grade III or higher toxicities occurred in 9 patients. Day 30 and day 100 mortalities were 3% and 15%, respectively. Patients achieved myeloid and platelet engraftment at a median of 14 days after transplantation. Long-term toxicities occurred in 2 patients: hypokalemia and tremor, both grade III, on days 370 and 361 after transplantation. Fourteen patients died, 7 of relapse-related causes and 7 of non-relapse-related causes. With a median follow-up for living patients of 14.7 months, 1-year overall survival, event-free survival, and non-relapse-related mortality were 75%, 65%, and 19%, respectively. Addition of TMLI to RIC is feasible and safe and could be offered to patients with advanced hematologic malignancies who might not otherwise be candidates for RIC.
- Published
- 2011
- Full Text
- View/download PDF
19. Reciprocal differentiation and tissue-specific pathogenesis of Th1, Th2, and Th17 cells in graft-versus-host disease.
- Author
-
Yi T, Chen Y, Wang L, Du G, Huang D, Zhao D, Johnston H, Young J, Todorov I, Umetsu DT, Chen L, Iwakura Y, Kandeel F, Forman S, and Zeng D
- Subjects
- Animals, Antibodies, Monoclonal immunology, B7-1 Antigen physiology, B7-H1 Antigen, CD4-Positive T-Lymphocytes immunology, CD4-Positive T-Lymphocytes pathology, Cell Movement, Flow Cytometry, Graft vs Host Disease metabolism, Idiopathic Interstitial Pneumonias etiology, Idiopathic Interstitial Pneumonias pathology, Interferon-gamma physiology, Interleukin-17 physiology, Interleukin-4, Lung immunology, Lung pathology, Membrane Glycoproteins physiology, Mice, Mice, Inbred BALB C, Mice, Inbred C57BL, Mice, Knockout, Neutralization Tests, Peptides physiology, Skin immunology, Skin pathology, T-Lymphocytes, Helper-Inducer immunology, Th1 Cells immunology, Th2 Cells immunology, Cell Differentiation, Graft vs Host Disease immunology, Graft vs Host Disease pathology, T-Lymphocytes, Helper-Inducer pathology, Th1 Cells pathology, Th2 Cells pathology
- Abstract
In acute graft-versus-host disease (GVHD), naive donor CD4(+) T cells recognize alloantigens on host antigen-presenting cells and differentiate into T helper (Th) subsets (Th1, Th2, and Th17 cells), but the role of Th subsets in GVHD pathogenesis is incompletely characterized. Here we report that, in an MHC-mismatched model of C57BL/6 donor to BALB/c recipient, WT donor CD4(+) T cells predominantly differentiated into Th1 cells and preferentially mediated GVHD tissue damage in gut and liver. However, absence of interferon-gamma (IFN-gamma) in CD4(+) T cells resulted in augmented Th2 and Th17 differentiation and exacerbated tissue damage in lung and skin; absence of both IL-4 and IFN-gamma resulted in augmented Th17 differentiation and preferential, although not exclusive, tissue damage in skin; and absence of both IFN-gamma and IL-17 led to further augmentation of Th2 differentiation and idiopathic pneumonia. The tissue-specific GVHD mediated by Th1, Th2, and Th17 cells was in part associated with their tissue-specific migration mediated by differential expression of chemokine receptors. Furthermore, lack of tissue expression of the IFN-gamma-inducible B7-H1 played a critical role in augmenting the Th2-mediated idiopathic pneumonia. These results indicate donor CD4(+) T cells can reciprocally differentiate into Th1, Th2, and Th17 cells that mediate organ-specific GVHD.
- Published
- 2009
- Full Text
- View/download PDF
20. Anti-CD3 preconditioning separates GVL from GVHD via modulating host dendritic cell and donor T-cell migration in recipients conditioned with TBI.
- Author
-
Li N, Chen Y, He W, Yi T, Zhao D, Zhang C, Lin CL, Todorov I, Kandeel F, Forman S, and Zeng D
- Subjects
- Animals, Antigens, CD immunology, Blood Donors, Cell Differentiation, Cell Movement, Cells, Cultured, Chemokines metabolism, Down-Regulation, Hematopoietic Stem Cell Transplantation, Integrin alpha Chains immunology, Leukemia pathology, Leukemia surgery, Mice, Receptors, Chemokine immunology, T-Lymphocytes metabolism, Transplantation Conditioning, Transplantation, Homologous, Up-Regulation, Whole-Body Irradiation, Antibodies immunology, CD3 Complex immunology, Dendritic Cells immunology, Graft vs Host Disease immunology, Leukemia immunology, T-Lymphocytes cytology, T-Lymphocytes immunology
- Abstract
Host dendritic cells (DCs) play a critical role in initiating graft-versus-host disease (GVHD) and graft-versus-leukemia (GVL), and separation of GVL from GVHD remains a major challenge in the treatment of hematologic malignancies by allogeneic hematopoietic cell transplantation (HCT). Here, we show that preconditioning with anti-CD3 monoclonal antibody before conditioning with total body irradiation (TBI) prevents GVHD but retains GVL in a HCT model of major histocompatibility complex (MHC)-mismatched C57BL/6 donor to BALB/c host. Prevention of GVHD is associated with inhibition of donor T-cell expression of homing and chemokine receptors, and inhibition of GVHD target tissue expression of chemokines. Furthermore, inhibition of donor T-cell expression of gut homing alpha4beta7 and chemokine receptor (CCR)9 by anti-CD3 preconditioning results from a reduction of CD103(+) DCs in draining mesenteric lymph nodes (LNs), which is associated with down-regulation of DC expression of CCR7, a receptor required for tissue DC migration to draining LNs. These results indicate that anti-CD3 preconditioning reduces not only tissue release of chemokines but also prevents tissue DC migration to draining LNs and subsequently reduces the capacity of DCs of draining LNs to imprint donor T-cell tissue tropism. Therefore, modulation of host DCs by anti-CD3 preconditioning before HCT represents a new approach for separating GVL from GVHD.
- Published
- 2009
- Full Text
- View/download PDF
21. Absence of donor Th17 leads to augmented Th1 differentiation and exacerbated acute graft-versus-host disease.
- Author
-
Yi T, Zhao D, Lin CL, Zhang C, Chen Y, Todorov I, LeBon T, Kandeel F, Forman S, and Zeng D
- Subjects
- Animals, Base Sequence, Cell Differentiation, Chemokines genetics, DNA Primers genetics, Gene Expression, Graft vs Host Disease genetics, Graft vs Host Disease immunology, Graft vs Host Disease pathology, Hematopoietic Stem Cell Transplantation adverse effects, Interferon-gamma antagonists & inhibitors, Interleukin-17 administration & dosage, Interleukin-17 biosynthesis, Interleukin-17 deficiency, Interleukin-17 genetics, Mice, Mice, Inbred BALB C, Mice, Inbred C57BL, Mice, Knockout, Neutralization Tests, T-Lymphocytes, Helper-Inducer classification, T-Lymphocytes, Helper-Inducer cytology, T-Lymphocytes, Helper-Inducer immunology, Tissue Donors, Transplantation, Homologous, Graft vs Host Disease etiology, T-Lymphocyte Subsets immunology, T-Lymphocyte Subsets pathology, Th1 Cells immunology, Th1 Cells pathology
- Abstract
Th17 is a newly identified T-cell lineage that secretes proinflammatory cytokine IL-17. Th17 cells have been shown to play a critical role in mediating autoimmune diseases such as EAE, colitis, and arthritis, but their role in the pathogenesis of graft-versus-host disease (GVHD) is still unknown. Here we showed that, in an acute GVHD model of C57BL/6 (H-2(b)) donor to BALB/c (H-2(d)) recipient, IL-17(-/-) donor T cells manifested an augmented Th1 differentiation and IFN-gamma production and induced exacerbated acute GVHD. Severe tissue damage mediated by IL-17(-/-) donor T cells was associated with increased Th1 infiltration, up-regulation of chemokine receptors by donor T cells, and enhanced tissue expression of inflammatory chemokines. Administration of recombinant IL-17 and neutralizing IFN-gamma in the recipients given IL-17(-/-) donor cells ameliorated the acute GVHD. Furthermore, the regulation of Th1 differentiation by IL-17 or Th17 may be through its influence on host DCs. Our results indicate that donor Th17 cells can down-regulate Th1 differentiation and ameliorate acute GVHD in allogeneic recipients, and that treatments neutralizing proinflammatory cytokine IL-17 may augment acute GVHD as well as other inflammatory autoimmune diseases.
- Published
- 2008
- Full Text
- View/download PDF
22. In vivo-activated CD103+CD4+ regulatory T cells ameliorate ongoing chronic graft-versus-host disease.
- Author
-
Zhao D, Zhang C, Yi T, Lin CL, Todorov I, Kandeel F, Forman S, and Zeng D
- Subjects
- Animals, Apoptosis immunology, Autoantibodies biosynthesis, Chronic Disease, Graft vs Host Disease immunology, Graft vs Host Disease pathology, Interleukin-2 Receptor alpha Subunit metabolism, Lymphocyte Activation, Male, Mice, Mice, Inbred BALB C, Mice, Inbred DBA, Plasma Cells immunology, Plasma Cells pathology, Spleen cytology, Spleen immunology, Spleen transplantation, Syndecans immunology, T-Lymphocytes, Regulatory classification, Tissue Donors, Transplantation, Homologous, Antigens, CD metabolism, Graft vs Host Disease therapy, Integrin alpha Chains metabolism, T-Lymphocytes, Regulatory immunology, T-Lymphocytes, Regulatory transplantation
- Abstract
CD103 (alphaEbeta7) has been shown to be an excellent marker for identifying in vivo-activated FoxP3(+)CD4(+) regulatory T (Treg) cells. It is unknown whether reinfusion of in vivo-activated donor-type CD103(+) Treg cells from recipient can ameliorate ongoing chronic graft-versus-host disease (GVHD). Here, we showed that, in a chronic GVHD model of DBA/2 (H-2(d)) donor to BALB/c (H-2(d)) recipient, donor-type CD103(+) Treg cells from recipients were much more potent than CD25(hi) natural Treg cells from donors in reversing clinical signs of GVHD and tissue damage. Furthermore, in contrast to CD25(hi) natural Treg cells, CD103(+) Treg cells expressed high levels of CCR5 but low levels of CD62L and directly migrated to GVHD target tissues. In addition, the CD103(+) Treg cells strongly suppressed donor CD4(+) T-cell proliferation; they also induced apoptosis of in vivo-activated CD4(+) T and B cells and significantly reduced pathogenic T and B cells in GVHD target tissues. These results indicate that CD103(+) Treg cells from chronic GVHD recipients are functional, and reinfusion of the CD103(+) Treg cells can shift the balance between Treg cells and pathogenic T cells in chronic GVHD recipients and ameliorate ongoing disease.
- Published
- 2008
- Full Text
- View/download PDF
23. The novel histone deacetylase inhibitor, LBH589, induces expression of DNA damage response genes and apoptosis in Ph- acute lymphoblastic leukemia cells.
- Author
-
Scuto A, Kirschbaum M, Kowolik C, Kretzner L, Juhasz A, Atadja P, Pullarkat V, Bhatia R, Forman S, Yen Y, and Jove R
- Subjects
- Acetylation, DNA Damage genetics, Gene Expression Regulation drug effects, Histones metabolism, Humans, Indoles, Intracellular Signaling Peptides and Proteins genetics, Panobinostat, Precursor Cell Lymphoblastic Leukemia-Lymphoma pathology, Tumor Cells, Cultured, Apoptosis drug effects, DNA Damage drug effects, Hydroxamic Acids pharmacology, Precursor Cell Lymphoblastic Leukemia-Lymphoma drug therapy
- Abstract
We investigated the mechanism of action of LBH589, a novel broad-spectrum HDAC inhibitor belonging to the hydroxamate class, in Philadelphia chromosome-negative (Ph(-)) acute lymphoblastic leukemia (ALL). Two model human Ph(-) ALL cell lines (T-cell MOLT-4 and pre-B-cell Reh) were treated with LBH589 and evaluated for biologic and gene expression responses. Low nanomolar concentrations (IC(50): 5-20 nM) of LBH589 induced cell-cycle arrest, apoptosis, and histone (H3K9 and H4K8) hyperacetylation. LBH589 treatment increased mRNA levels of proapoptosis, growth arrest, and DNA damage repair genes including FANCG, FOXO3A, GADD45A, GADD45B, and GADD45G. The most dramatically expressed gene (up to 45-fold induction) observed after treatment with LBH589 is GADD45G. LBH589 treatment was associated with increased histone acetylation at the GADD45G promoter and phosphorylation of histone H2A.X. Furthermore, treatment with LBH589 was active against cultured primary Ph(-) ALL cells, including those from a relapsed patient, inducing loss of cell viability (up to 70%) and induction of GADD45G mRNA expression (up to 35-fold). Thus, LBH589 possesses potent growth inhibitory activity against including Ph(-) ALL cells associated with up-regulation of genes critical for DNA damage response and growth arrest. These findings provide a rationale for exploring the clinical activity of LBH589 in the treatment of patients with Ph(-) ALL.
- Published
- 2008
- Full Text
- View/download PDF
24. Diabetes, hypertension, and cardiovascular events in survivors of hematopoietic cell transplantation: a report from the bone marrow transplantation survivor study.
- Author
-
Baker KS, Ness KK, Steinberger J, Carter A, Francisco L, Burns LJ, Sklar C, Forman S, Weisdorf D, Gurney JG, and Bhatia S
- Subjects
- Adult, Cardiovascular Diseases epidemiology, Case-Control Studies, Data Collection, Diabetes Mellitus epidemiology, Follow-Up Studies, Humans, Hypertension epidemiology, Prevalence, Siblings, Survivors, Transplantation, Autologous, Transplantation, Homologous, Whole-Body Irradiation adverse effects, Cardiovascular Diseases etiology, Diabetes Mellitus etiology, Hematopoietic Stem Cell Transplantation adverse effects, Hypertension etiology
- Abstract
We ascertained the prevalence of self-reported late occurrence of diabetes, hypertension, and cardiovascular (CV) disease in 1089 hematopoietic cell transplantation (HCT) survivors who underwent HCT between 1974 and 1998, survived at least 2 years, and were not currently taking immunosuppressant agents and compared them with 383 sibling controls. All subjects completed a 255-item health questionnaire. The mean age at survey completion was 39.3 years for survivors and 38.6 years for siblings; mean follow-up was 8.6 years. Adjusting for age, sex, race, and body mass index (BMI), survivors of allogeneic HCT were 3.65 times (95% confidence interval [CI], 1.82-7.32) more likely to report diabetes than siblings and 2.06 times (95% CI, 1.39-3.04) more likely to report hypertension compared with siblings but did not report other CV outcomes with any greater frequency. Recipients of autologous HCTs were no more likely than siblings to report any of the outcomes studied. Allogeneic HCT survivors were also more likely to develop hypertension (odds ratio [OR]=2.31; 95% CI, 1.45-3.67) than autologous recipients. Total body irradiation (TBI) exposure was associated with an increased risk of diabetes (OR=3.42; 95% CI, 1.55-7.52). Thus, HCT survivors have a higher age- and BMI-adjusted risk of diabetes and hypertension, potentially leading to a higher than expected risk of CV events with age.
- Published
- 2007
- Full Text
- View/download PDF
25. Impact of chronic graft-versus-host disease on the health status of hematopoietic cell transplantation survivors: a report from the Bone Marrow Transplant Survivor Study.
- Author
-
Fraser CJ, Bhatia S, Ness K, Carter A, Francisco L, Arora M, Parker P, Forman S, Weisdorf D, Gurney JG, and Baker KS
- Subjects
- Adult, California, Chronic Disease, Female, Graft vs Host Disease physiopathology, Graft vs Host Disease psychology, Health Status, Hematopoietic Stem Cell Transplantation psychology, Humans, Male, Middle Aged, Minnesota, Odds Ratio, Quality of Life, Surveys and Questionnaires, Time Factors, Transplantation, Homologous, Graft vs Host Disease etiology, Hematopoietic Stem Cell Transplantation adverse effects
- Abstract
The aim of this study was to understand the impact of chronic graft-versus-host disease (cGVHD) on the overall health status of hematopoietic cell transplantation (HCT) survivors. Subjects included 584 individuals who had undergone allogeneic HCT between 1976 and 1999, survived 2 or more years, and completed a 255-item health questionnaire. Global assessment of health status was facilitated by measurement of 6 health status domains: general health, mental health, functional impairment, activity limitation, pain, and anxiety/fear. Information regarding diagnosis of cGVHD was abstracted from medical records, and presence of active cGVHD in the preceding 12 months was self-reported. The incidence of cGVHD in participants was 54%, of whom 46% reported active cGVHD. In multivariable analyses, subjects with active cGVHD were more likely to report adverse general health, mental health, functional impairments, activity limitation, and pain than were those with no history of cGVHD. However, health status did not differ between those with resolved cGVHD and those who never had cGVHD. We conclude that active cGVHD has a significant impact on many aspects of the overall health status of HCT survivors and that, most importantly, those successfully treated for cGVHD do not appear to have long-term impairments.
- Published
- 2006
- Full Text
- View/download PDF
26. Donor CD4+ T and B cells in transplants induce chronic graft-versus-host disease with autoimmune manifestations.
- Author
-
Zhang C, Todorov I, Zhang Z, Liu Y, Kandeel F, Forman S, Strober S, and Zeng D
- Subjects
- Animals, Autoimmune Diseases blood, Autoimmune Diseases immunology, Autoimmune Diseases pathology, Disease Models, Animal, Graft vs Host Disease blood, Graft vs Host Disease pathology, Lymphocyte Activation, Male, Mice, Mice, Inbred BALB C, Mice, Inbred DBA, Spleen immunology, Transplantation, Homologous, Autoimmune Diseases therapy, B-Lymphocytes transplantation, CD4-Positive T-Lymphocytes transplantation, Graft vs Host Disease immunology, Lymphocyte Transfusion adverse effects, Stem Cell Transplantation adverse effects
- Abstract
Chronic graft-vs-host disease (GVHD) is a major cause of morbidity and mortality of long-term survivors of allogeneic hemato-poietic cell transplantation (HCT). Chronic GVHD can have features of an autoimmune collagen vascular disease with clinical manifestations similar to autoimmune scleroderma and systemic lupus erythematosus (SLE). However, the pathogenesis of chronic GVHD is poorly understood. It is unclear how autoreactive T and B cells are generated in chronic GVHD recipients. We have recently developed a new chronic GVHD model by transplantation of donor DBA/2 (H-2d) spleen cells into major histocompatibility complex (MHC)-matched but minor antigen-mismatched sublethally irradiated BALB/c (H-2d) recipients as well as athymic BALB/c(nu/nu) and adult-thymectomized BALB/c recipients. Both euthymic and athymic BALB/c recipients developed high levels of serum IgG autoantibodies, sclerodermatous skin damage, and glomerulonephritis. Disease induction required both donor CD25-CD4+ T and B cells in transplants. In contrast, donor CD25+CD4+ T regulatory (Treg) cells prevented the disease induction. These results indicate that host thymus is not required for induction of chronic GVHD and that quiescent autoreactive T and B cells in transplants from nonautoimmune donors may be activated and expanded to cause chronic GVHD with autoimmune manifestations in allogeneic recipients, and donor Treg cells can suppress this process.
- Published
- 2006
- Full Text
- View/download PDF
27. A phase 1/2 trial of high-dose yttrium-90-ibritumomab tiuxetan in combination with high-dose etoposide and cyclophosphamide followed by autologous stem cell transplantation in patients with poor-risk or relapsed non-Hodgkin lymphoma.
- Author
-
Nademanee A, Forman S, Molina A, Fung H, Smith D, Dagis A, Kwok C, Yamauchi D, Anderson AL, Falk P, Krishnan A, Kirschbaum M, Kogut N, Nakamura R, O'donnell M, Parker P, Popplewell L, Pullarkat V, Rodriguez R, Sahebi F, Smith E, Snyder D, Stein A, Spielberger R, Zain J, White C, and Raubitschek A
- Subjects
- Adult, Antibodies, Monoclonal administration & dosage, Antibodies, Monoclonal adverse effects, Cyclophosphamide administration & dosage, Cyclophosphamide adverse effects, Drug Therapy, Combination, Etoposide administration & dosage, Etoposide adverse effects, Female, Humans, Lymphoma, Non-Hodgkin immunology, Lymphoma, Non-Hodgkin surgery, Male, Middle Aged, Positron-Emission Tomography, Prognosis, Recurrence, Survival Rate, Transplantation, Autologous, Yttrium Radioisotopes administration & dosage, Yttrium Radioisotopes adverse effects, Yttrium Radioisotopes therapeutic use, Antibodies, Monoclonal therapeutic use, Cyclophosphamide therapeutic use, Etoposide therapeutic use, Lymphoma, Non-Hodgkin diagnosis, Lymphoma, Non-Hodgkin therapy, Radioimmunotherapy, Stem Cell Transplantation
- Abstract
We conducted a phase 1/2 trial of high-dose 90Y-ibritumomab tiuxetan in combination with high-dose etoposide (VP-16) 40 to 60 mg/kg (day -4) and cyclophosphamide 100 mg/kg (day -2) followed by autologous stem cell transplantation (ASCT) in 31 patients with CD20+ non-Hodgkin lymphoma (NHL). Patients underwent dosimetry (day -21) with 5 mCi (185 MBq) 111In-ibritumomab tiuxetan following 250 mg/m2 rituximab, followed a week later by 90Y-ibritumomab tiuxetan to deliver a target dose of 1000 cGy to highest normal organ. Bone marrow biopsy was done on day -7 to estimate radiation dose and stem cells were reinfused when the radiation dose was estimated to be less than 5 cGy. The median 90Y-ibritumomab tiuxetan dose was 71.6 mCi (2649.2 MBq; range, 36.6-105 mCi; range, 1354.2-3885 MBq). Histology included follicular lymphoma (n = 12), diffuse large B-cell (n = 14), and mantle cell (n = 5). The median number of prior chemo-therapy treatments was 2. The treatment was well tolerated. The median times to reach an absolute neutrophil count greater than 500/microL and platelet count more than 20,000/microL were 10 days and 12 days, respectively. There were 2 deaths and 5 relapses. At a median follow-up of 22 months, the 2-year estimated overall survival and relapse-free survival rates are 92% and 78%, respectively. We conclude that high-dose 90Y-ibritumomab tiuxetan can be combined safely with high-dose etoposide and cyclophosphamide without an increase in transplant-related toxicity or delayed engraftment.
- Published
- 2005
- Full Text
- View/download PDF
28. An anti-CD20-IL-2 immunocytokine is highly efficacious in a SCID mouse model of established human B lymphoma.
- Author
-
Gillies SD, Lan Y, Williams S, Carr F, Forman S, Raubitschek A, and Lo KM
- Subjects
- Animals, Antibodies immunology, Antibodies, Monoclonal immunology, Antibodies, Monoclonal therapeutic use, Antibodies, Monoclonal, Murine-Derived, Apoptosis, Cell Line, Tumor, Humans, Mice, Mice, Inbred BALB C, Mice, SCID, Neoplasm Metastasis, Neoplasm, Residual drug therapy, Neoplasm, Residual immunology, Rituximab, Survival Rate, Xenograft Model Antitumor Assays, Antigens, CD20 immunology, Disease Models, Animal, Immunotherapy, Interleukin-2 immunology, Lymphoma, B-Cell drug therapy, Lymphoma, B-Cell immunology
- Abstract
We have engineered an anti-CD20-interleukin 2 (IL-2) immunocytokine (ICK) based on the Leu16 anti-CD20 antibody and have deimmunized both the variable (V) regions as well as the junction between the heavy (H) chain constant region and IL-2. Mutations were made to remove potential T-cell epitopes identified by in silico binding to major histocompatibility complex (MHC) class II molecules. The resulting immunocytokine, DI-Leu16-IL-2, retained full anti-CD20 activity as assessed by fluorescence-activated cell-sorting (FACS) analysis, and had enhanced antibody-dependent cellular cytotoxicity (ADCC) effector function relative to the DI-Leu16 antibody or control anti-CD20 antibody (rituximab). In a severe combined immunodeficient (SCID) mouse model of disseminated, residual lymphoma, anti-CD20-IL-2 immunocytokines based on Leu16 were far more effective at a dose of 0.25 mg/kg than anti-CD20 antibody given at 25/mg/kg, despite a shorter half-life of the ICK. Anti-CD20-IL-2 was also far more effective than a control ICK targeted to an antigen with greatly reduced expression on Daudi tumor cells, or various combinations of anti-CD20 antibodies and IL-2. Antitumor activity of DI-Leu16-IL-2 was shown to partially but not entirely depend on Fc receptor (R) binding, suggesting that ADCC and targeting of IL-2 both play roles in the mechanism of tumor clearance. Based on these animal models, DI-Leu16-IL-2 could offer therapeutic potential for patients with CD20 positive lymphoma. Clinical trials are currently under development.
- Published
- 2005
- Full Text
- View/download PDF
29. Donor CD8+ T cells facilitate induction of chimerism and tolerance without GVHD in autoimmune NOD mice conditioned with anti-CD3 mAb.
- Author
-
Liang Y, Huang T, Zhang C, Todorov I, Atkinson M, Kandeel F, Forman S, and Zeng D
- Subjects
- Animals, Antibodies, Monoclonal therapeutic use, CD3 Complex immunology, Cell Proliferation, Cytokines biosynthesis, Mice, Mice, Inbred NOD, Transplantation Conditioning methods, Bone Marrow Transplantation methods, CD8-Positive T-Lymphocytes transplantation, Diabetes Mellitus, Type 1 therapy, Graft vs Host Disease prevention & control, Immune Tolerance, Transplantation Chimera
- Abstract
Prevention of autoimmune diabetes and induction of islet transplantation tolerance in nonobese diabetic (NOD) mice can be reached by induction of mixed chimerism via bone marrow transplantation (BMT), but this procedure requires total body irradiation (TBI) conditioning of the recipients. The toxicity of radiation and potential for graft-versus-host disease (GVHD) prevents its clinical application. Donor CD8+ T cells play a critical role in facilitation of engraftment but also contribute to induction of GVHD in TBI-conditioned recipients. Here, we showed that high doses of donor CD8+ T cells in combination with bone marrow (BM) cells induced mixed chimerism without GVHD in NOD recipients conditioned with anti-CD3 monoclonal antibody (mAb). The prevention of GVHD in those recipients was associated with low-level production of inflammatory cytokines (ie, tumor necrosis factor alpha [TNF-alpha]), high-level production of anti-inflammatory cytokines (ie, interleukin 4 [IL-4] and IL-10), and confining of the donor CD8+ T-cell expansion to lymphohematopoietic tissues. The chimeric NOD recipients showed donor-specific tolerance and reversal of insulitis. These results demonstrate that donor CD8+ T-cell-mediated facilitation of engraftment can be separated from GVHD in nonirradiated recipients. This regimen may have potential application in the treatment of autoimmune disorders as well as induction of transplantation tolerance.
- Published
- 2005
- Full Text
- View/download PDF
30. Autologous stem cell transplantation for HIV-associated lymphoma.
- Author
-
Krishnan A, Molina A, Zaia J, Nademanee A, Kogut N, Rosenthal J, Woo D, and Forman SJ
- Subjects
- Adolescent, Adult, Antiretroviral Therapy, Highly Active, Bacterial Infections complications, CD4 Lymphocyte Count, Child, Disease-Free Survival, Female, HIV Infections drug therapy, HIV Infections immunology, Humans, Lymphoma, AIDS-Related immunology, Lymphoma, AIDS-Related mortality, Male, Middle Aged, Neutropenia complications, Opportunistic Infections complications, Prognosis, Remission Induction, Transplantation Conditioning adverse effects, Transplantation, Autologous, Hematopoietic Stem Cell Transplantation adverse effects, Lymphoma, AIDS-Related therapy
- Abstract
Is peripheral stem cell mobilization followed by autologous stem cell transplantation (ASCT) feasible in patients with human immunodeficiency virus (HIV)- associated lymphoma (HIV-L)? Studies have demonstrated that, in the HIV- negative (HIV(-)) setting, ASCT may improve lymphoma-free survival in high-risk non-Hodgkin lymphoma (NHL) or relapsed Hodgkin disease (HD) and NHL. Given the poor prognosis of HIV-L with conventional chemotherapy, this dose-intensive approach was explored. Nine patients with HIV-HD or NHL mobilized a median of 10.6 x 10(6) CD34(+) cells/kg and engrafted after ASCT. CD4 counts recovered to pretransplantation levels and HIV viral loads were controlled in patients compliant with antiretroviral therapy. Seven of 9 patients remain in remission from their lymphoma at a median of 19 months after transplantation. Thus, patients with HIV-L on antiretroviral therapy can engraft following ASCT. Prolonged lymphoma remissions, without significant compromise of immune function, can be seen, suggesting that ASCT can be used in selected patients with HIV-L.
- Published
- 2001
- Full Text
- View/download PDF
31. Identification of a candidate human neurohematopoietic stem-cell population.
- Author
-
Shih CC, Weng Y, Mamelak A, LeBon T, Hu MC, and Forman SJ
- Subjects
- Abortion, Legal, Brain cytology, Cell Culture Techniques methods, Cell Division, Cells, Cultured, Cerebral Cortex embryology, Culture Media, Epidermal Growth Factor analysis, Female, Fetus, Fibroblast Growth Factor 2 analysis, Hematopoiesis, Humans, Immunohistochemistry, Pregnancy, Thymus Gland cytology, Thymus Gland embryology, Brain embryology, Cell Differentiation physiology, Cerebral Cortex cytology, Hematopoietic Stem Cells physiology
- Abstract
It was recently reported that transplantation of clonally derived murine neurosphere cells into sublethally irradiated allogeneic hosts leads to a donor-derived hematopoietic reconstitution. The confirmation of the existence of a common neurohematopoietic stem cell in the human brain will have a significant effect on stem cell research and on clinical transplantation. Here, it is demonstrated that the human fetal brain contains separate but overlapping epidermal growth factor (EGF)-responsive and basic fibroblast growth factor (FGF-2)-responsive neural stem cells. The majority (> 85%) of cells within these EGF- and/or FGF-2-generated neurospheres express characteristic neural stem/progenitor cell markers including nestin, EGF receptor, and FGF-2 receptor. These neural stem cells can be continuously passaged in vitro, and demonstrate a constant 20-fold expansion in every passage for up to the fifth passage (the longest period that has been carried out in the authors' laboratory). These neural stem cells are multipotential for neurons, astrocytes, and oligodendrocytes. After transplantation into SCID-hu mice, all neural stem cells, regardless of passages, culture conditions, and donors, are able to establish long-term hematopoietic reconstitution in the presence of an intact human bone marrow microenvironment.
- Published
- 2001
- Full Text
- View/download PDF
32. Karyotypic analysis predicts outcome of preremission and postremission therapy in adult acute myeloid leukemia: a Southwest Oncology Group/Eastern Cooperative Oncology Group Study.
- Author
-
Slovak ML, Kopecky KJ, Cassileth PA, Harrington DH, Theil KS, Mohamed A, Paietta E, Willman CL, Head DR, Rowe JM, Forman SJ, and Appelbaum FR
- Subjects
- Acute Disease, Adolescent, Adult, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Bone Marrow Transplantation, Chromosomes, Human ultrastructure, Combined Modality Therapy, Cytarabine administration & dosage, Female, Humans, Idarubicin administration & dosage, Leukemia, Myeloid drug therapy, Leukemia, Myeloid mortality, Leukemia, Myeloid therapy, Life Tables, Male, Middle Aged, Remission Induction, Risk, Survival Analysis, Translocation, Genetic, Transplantation, Autologous, Transplantation, Homologous, Treatment Outcome, Aneuploidy, Chromosome Aberrations, Karyotyping, Leukemia, Myeloid genetics
- Abstract
The associations of cytogenetics with complete remission (CR) rates, overall survival (OS), and outcomes after CR were studied in 609 previously untreated AML patients younger than 56 years old in a clinical trial comparing 3 intensive postremission therapies: intensive chemotherapy, autologous transplantation (ABMT), or allogeneic bone marrow transplantation (alloBMT) from matched related donors. Patients were categorized into favorable, intermediate, unfavorable, and unknown cytogenetic risk groups based on pretreatment karyotypes. CR rates varied significantly (P <.0001) among the 4 groups: favorable, 84% (95% confidence interval [CI], 77%-90%); intermediate, 76% (CI, 71%-81%); unfavorable, 55% (CI, 48%-63%); and unknown, 54% (CI, 33%-74%). There was similar significant heterogeneity of OS (P <.0001), with the estimated relative risk of death from any cause being 1.50 (CI, 1.10-2.05), 3. 33 (CI, 2.43-4.55), and 2.66 (CI, 1.59-4.45) for the intermediate, unfavorable, and unknown risk groups, respectively, compared with the favorable group. In multivariate analyses, the effects of cytogenetic risk status on CR rate and OS could not be explained by other patient or disease characteristics. Among postremission patients, survival from CR varied significantly among favorable, intermediate, and unfavorable groups (P =.0003), with significant evidence of interaction (P =.017) between the effects of treatment and cytogenetic risk status on survival. Patients with favorable cytogenetics did significantly better following ABMT and alloBMT than with chemotherapy alone, whereas patients with unfavorable cytogenetics did better with alloBMT. Cytogenetic risk status is a significant factor in predicting response of AML patients to therapy; however, to tighten treatment correlates within genetically defined AML subsets, a significantly larger leukemia cytogenetic database is warranted.
- Published
- 2000
33. A secreted and LIF-mediated stromal cell-derived activity that promotes ex vivo expansion of human hematopoietic stem cells.
- Author
-
Shih CC, Hu MC, Hu J, Weng Y, Yazaki PJ, Medeiros J, and Forman SJ
- Subjects
- Animals, Antigens, CD34 metabolism, Bone Marrow embryology, Cell Differentiation, Cytokines metabolism, Enzyme-Linked Immunosorbent Assay, Growth Inhibitors pharmacology, Humans, Leukemia Inhibitory Factor, Lymphokines pharmacology, Mice, Mice, SCID, Phenotype, Thy-1 Antigens metabolism, Cell Culture Techniques methods, Culture Media, Conditioned metabolism, Growth Inhibitors metabolism, Hematopoietic Stem Cell Transplantation, Hematopoietic Stem Cells cytology, Interleukin-6, Lymphokines metabolism, Stromal Cells metabolism
- Abstract
The development of culture systems that facilitate ex vivo maintenance and expansion of transplantable hematopoietic stem cells (HSCs) is vital to stem cell research. Establishment of such culture systems will have significant impact on ex vivo manipulation and expansion of transplantable stem cells in clinical applications such as gene therapy, tumor cell purging, and stem cell transplantation. We have recently developed a stromal-based culture system that facilitates ex vivo expansion of transplantable human HSCs. In this stromal-based culture system, 2 major contributors to the ex vivo stem cell expansion are the addition of leukemia inhibitory factor (LIF) and the AC6.21 stromal cells. Because the action of LIF is indirect and mediated by stromal cells, we hypothesized that LIF binds to the LIF receptor on AC6.21 stromal cells, leading to up-regulated production of stem cell expansion promoting factor (SCEPF) and/or down-regulated production of stem cell expansion inhibitory factor (SCEIF). Here we demonstrate a secreted SCEPF activity in the conditioned media of LIF-treated AC6.21 stromal cell cultures (SCM-LIF). The magnitude of ex vivo stem cell expansion depends on the concentration of the secreted SCEPF activity in the SCM-LIF. Furthermore, we have ruled out the contribution of 6 known early-acting cytokines, including interleukin-3, interleukin-6, granulocyte macrophage colony-stimulating factor, stem cell factor, flt3 ligand, and thrombopoietin, to this SCEPF activity. Although further studies are required to characterize this secreted SCEPF activity and to determine whether this secreted SCEPF activity is mediated by a single factor or by multiple growth factors, our results demonstrate that stromal cells are not required for this secreted SCEPF activity to facilitate ex vivo stem cell expansion. (Blood. 2000;95:1957-1966)
- Published
- 2000
34. Predictors of therapy-related leukemia and myelodysplasia following autologous transplantation for lymphoma: an assessment of risk factors.
- Author
-
Krishnan A, Bhatia S, Slovak ML, Arber DA, Niland JC, Nademanee A, Fung H, Bhatia R, Kashyap A, Molina A, O'Donnell MR, Parker PA, Sniecinski I, Snyder DS, Spielberger R, Stein A, and Forman SJ
- Subjects
- Adolescent, Adult, Aged, Antineoplastic Combined Chemotherapy Protocols administration & dosage, Antineoplastic Combined Chemotherapy Protocols adverse effects, Antineoplastic Combined Chemotherapy Protocols therapeutic use, California epidemiology, Case-Control Studies, Cohort Studies, Combined Modality Therapy, Etoposide administration & dosage, Etoposide adverse effects, Female, Hematopoietic Stem Cell Mobilization adverse effects, Hematopoietic Stem Cell Mobilization methods, Humans, Leukemia chemically induced, Leukemia epidemiology, Leukemia, Radiation-Induced epidemiology, Leukemia, Radiation-Induced etiology, Lymphoma drug therapy, Lymphoma radiotherapy, Male, Middle Aged, Multivariate Analysis, Myelodysplastic Syndromes chemically induced, Myelodysplastic Syndromes epidemiology, Neoplasms, Second Primary chemically induced, Neoplasms, Second Primary epidemiology, Odds Ratio, Radiotherapy adverse effects, Retrospective Studies, Risk Factors, Treatment Outcome, Whole-Body Irradiation adverse effects, Hematopoietic Stem Cell Transplantation adverse effects, Leukemia etiology, Lymphoma therapy, Myelodysplastic Syndromes etiology, Neoplasms, Second Primary etiology, Transplantation Conditioning adverse effects
- Abstract
We analyzed data on 612 patients who had undergone high-dose chemoradiotherapy (HDT) with autologous stem cell rescue for Hodgkin's disease (HD) and non-Hodgkin's lymphoma (NHL) at the City of Hope National Medical Center, to evaluate the incidence of therapy-related myelodysplasia (t-MDS) or therapy-related acute myeloid leukemia (t-AML) and associated risk factors. A retrospective cohort and a nested case-control study design were used to evaluate the role of pretransplant therapeutic exposures and transplant conditioning regimens. Twenty-two patients developed morphologic evidence of t-MDS/t-AML. The estimated cumulative probability of developing morphologic t-MDS/t-AML was 8.6% +/- 2.1% at 6 years. Multivariate analysis of the entire cohort revealed stem cell priming with VP-16 (RR = 7.7, P = 0.002) to be independently associated with an increased risk of t-MDS/t-AML. The influence of pretransplant therapy on subsequent t-MDS/t-AML risk was determined by a case-control study. Multivariate analysis revealed an association between pretransplant radiation and the risk of t-MDS/t-AML, but failed to reveal any association with pretransplant chemotherapy or conditioning regimens. However, patients who had been primed with VP-16 for stem cell mobilization were at a 12. 3-fold increased risk of developing t-AML with 11q23/21q22 abnormalities (P = 0.006). Patients undergoing HDT with stem cell rescue are at an increased risk of t-MDS/t-AML, especially those receiving priming with VP-16 for peripheral stem cell collection. (Blood. 2000;95:1588-1593)
- Published
- 2000
35. Long-term ex vivo maintenance and expansion of transplantable human hematopoietic stem cells.
- Author
-
Shih CC, Hu MC, Hu J, Medeiros J, and Forman SJ
- Subjects
- Animals, Granulocyte-Macrophage Colony-Stimulating Factor pharmacology, Growth Inhibitors pharmacology, Humans, Interleukin-3 pharmacology, Interleukin-6 pharmacology, Leukemia Inhibitory Factor, Lymphokines pharmacology, Mice, Mice, SCID, Cell Culture Techniques methods, Hematopoietic Stem Cell Mobilization, Hematopoietic Stem Cell Transplantation, Hematopoietic Stem Cells cytology
- Abstract
We have developed a stromal-based in vitro culture system that facilitates ex vivo expansion of transplantable CD34(+) thy-1(+) cells using long-term hematopoietic reconstitution in severe combined immunodeficient-human (SCID-hu) mice as an in vivo assay for transplantable human hematopoietic stem cells (HSCs). The addition of leukemia inhibitory factor (LIF) to purified CD34(+) thy-1(+) cells on AC6.21 stroma, a murine bone marrow-derived stromal cell line, caused expansion of cells with CD34(+) thy-1(+) phenotype. Addition of other cytokines, including interleukin-3 (IL-3), IL-6, granulocyte-macrophage colony-stimulating factor, and stem cell factor, to LIF in the cultures caused a 150-fold expansion of cells retaining the CD34(+) thy-1(+) phenotype. The ex vivo-expanded CD34(+) thy-1(+) cells gave rise to multilineage differentiation, including myeloid, T, and B cells, when transplanted into SCID-hu mice. Both murine LIF (cannot bind to human LIF receptor) and human LIF caused expansion of human CD34(+) thy-1(+) cells in vitro, suggesting action through the murine stroma. Furthermore, another human HSC candidate, CD34(+) CD38(-) cells, shows a similar pattern of proliferative response. This suggests that ex vivo expansion of transplantable human stem cells under this in vitro culture system is a general phenomenon and not just specific for CD34(+) thy-1(+) cells.
- Published
- 1999
36. Selective ablation of acute myeloid leukemia using antibody-targeted chemotherapy: a phase I study of an anti-CD33 calicheamicin immunoconjugate.
- Author
-
Sievers EL, Appelbaum FR, Spielberger RT, Forman SJ, Flowers D, Smith FO, Shannon-Dorcy K, Berger MS, and Bernstein ID
- Subjects
- Adult, Aged, Blood Cell Count drug effects, Enediynes, Female, Hematopoiesis drug effects, Humans, Immunoconjugates immunology, Injections, Intravenous, Leukemia, Myeloid immunology, Leukemia, Myeloid physiopathology, Male, Middle Aged, Sialic Acid Binding Ig-like Lectin 3, Treatment Outcome, Aminoglycosides, Anti-Bacterial Agents administration & dosage, Antibiotics, Antineoplastic administration & dosage, Antibodies, Monoclonal administration & dosage, Antigens, CD immunology, Antigens, Differentiation, Myelomonocytic immunology, Immunoconjugates administration & dosage, Leukemia, Myeloid drug therapy
- Abstract
Leukemic blast cells express the CD33 antigen in most patients with acute myeloid leukemia (AML), but this antigen is not expressed by hematopoietic stem cells. We conducted a study to determine whether normal hematopoiesis could be restored in patients with AML by selective ablation of cells expressing the CD33 antigen. In a dose escalation study, 40 patients with relapsed or refractory CD33(+) AML were treated with an immunoconjugate (CMA-676) consisting of humanized anti-CD33 antibody linked to the potent antitumor antibiotic calicheamicin. The capacity of leukemic cells to efflux 3, 3'-diethyloxacarbocyanine iodide (DiOC2) was used to estimate pretreatment functional drug resistance. Leukemia was eliminated from the blood and marrow of 8 (20%) of the 40 patients; blood counts returned to normal in three (8%) patients. A high rate of clinical response was observed in leukemias characterized by low dye efflux in vitro. Infusions of CMA-676 were generally well tolerated, and a postinfusion syndrome of fever and chills was the most common toxic effect. Two patients who were treated at the highest dose level (9 mg/m2) were neutropenic >5 weeks after the last dose of CMA-676. These results show that an immunoconjugate targeted to CD33 can selectively ablate malignant hematopoiesis in some patients with AML.
- Published
- 1999
37. Recombinant human thrombopoietin in combination with granulocyte colony-stimulating factor enhances mobilization of peripheral blood progenitor cells, increases peripheral blood platelet concentration, and accelerates hematopoietic recovery following high-dose chemotherapy.
- Author
-
Somlo G, Sniecinski I, ter Veer A, Longmate J, Knutson G, Vuk-Pavlovic S, Bhatia R, Chow W, Leong L, Morgan R, Margolin K, Raschko J, Shibata S, Tetef M, Yen Y, Forman S, Jones D, Ashby M, Fyfe G, Hellmann S, and Doroshow JH
- Subjects
- Adult, Antineoplastic Combined Chemotherapy Protocols adverse effects, Blood Component Removal, Blood Platelets drug effects, Cisplatin administration & dosage, Cyclophosphamide administration & dosage, Etoposide administration & dosage, Female, Filgrastim, Granulocyte Colony-Stimulating Factor adverse effects, Hematopoietic Stem Cells drug effects, Humans, Middle Aged, Neoplasm Staging, Platelet Count drug effects, Recombinant Proteins adverse effects, Recombinant Proteins therapeutic use, Thrombopoietin adverse effects, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Blood Platelets physiology, Breast Neoplasms blood, Breast Neoplasms therapy, Granulocyte Colony-Stimulating Factor therapeutic use, Hematopoiesis, Hematopoietic Stem Cells physiology, Thrombopoietin therapeutic use
- Abstract
Lineage-specific growth factors mobilize peripheral blood progenitor cells (PBPC) and accelerate hematopoietic recovery after high-dose chemotherapy. Recombinant human thrombopoietin (rhTPO) may further increase the progenitor-cell content and regenerating potential of PBPC products. We evaluated the safety and activity of rhTPO as a PBPC mobilizer in combination with granulocyte colony-stimulating factor (G-CSF) in 29 breast cancer patients treated with high-dose chemotherapy followed by PBPC reinfusion. Initially, patients received escalating single doses of rhTPO intravenously (IV) at 0.6, 1.2, or 2.4 micrograms/kg, on day 1. Subsequent patients received rhTPO 0.6 or 0.3 micrograms/kg on days -3, -1, and 1, or 0.6 micrograms/kg on days -1 and 1. G-CSF, 5 micrograms/kg IV or subcutaneously (SC) twice daily, was started on day 3 and continued through aphereses. Twenty comparable, concurrently and identically treated patients (who were eligible and would have been treated on protocol but for the lack of study opening) mobilized with G-CSF alone served as comparisons. CD34(+) cell yields were substantially higher with the first apheresis following rhTPO and G-CSF versus G-CSF alone: 4.1 x 10(6)/kg (range, 1.3 to 17.6) versus 0.8 x 10(6)/ kg (range, 0.3 to 4.2), P =.0003. The targeted minimum yield of 3 x 10(6) CD34(+) cells/kg was procured following a single apheresis procedure in 61% of the rhTPO and G-CSF-mobilized group versus 10% of G-CSF-mobilized patients (P =.001). In rhTPO and G-CSF mobilized patients, granulocyte (day 8 v 9, P =.0001) and platelet recovery (day 9 v 10, P =.07) were accelerated, and fewer erythrocyte (3 v 4, P =.02) and platelet (4 v 5, P =.02) transfusions were needed compared with G-CSF-mobilized patients. Peripheral blood platelet counts, following rhTPO and G-CSF, were increased by greater than 100% and the platelet content of PBPC products by 60% to 110% on the first and second days of aphereses (P <.0001) with the greatest effect seen with repeated dosing of rhTPO at 0.6 microgram/kg. rhTPO is safe and well tolerated as a mobilizing agent before PBPC collection. Mobilization with rhTPO and G-CSF, in comparison to a comparable, nonrandomized G-CSF-mobilized group of patients, decreases the number of apheresis procedures required, may accelerate hematopoietic recovery, and may reduce the number of transfusions required following high-dose chemotherapy for breast cancer.
- Published
- 1999
38. Transduction of primitive human marrow and cord blood-derived hematopoietic progenitor cells with adeno-associated virus vectors.
- Author
-
Chatterjee S, Li W, Wong CA, Fisher-Adams G, Lu D, Guha M, Macer JA, Forman SJ, and Wong KK Jr
- Subjects
- Alkaline Phosphatase genetics, Antigens, CD34 analysis, Cells, Cultured, Gene Expression, Granulocytes, HIV Long Terminal Repeat genetics, Humans, In Situ Hybridization, Fluorescence, Macrophages, Placenta enzymology, RNA, Antisense, Time Factors, Transcription, Genetic, Bone Marrow Cells metabolism, Dependovirus genetics, Gene Transfer Techniques, Genetic Vectors, Hematopoietic Stem Cells metabolism
- Abstract
We evaluated the capacity of adeno-associated virus (AAV) vectors to transduce primitive human myeloid progenitor cells derived from marrow and cord blood in long-term cultures and long-term culture-initiating cell (LTC-IC) assays. Single-colony analyses showed that AAV vectors transduced CD34(+) and CD34(+)38(-) clonogenic cells in long-term culture. Gene transfer was readily observed in LTC-ICs derived from 5-, 8-, and 10-week cultures. Recombinant AAV (rAAV) transduction was observed in every donor analyzed, although a wide range of gene transfer frequencies (5% to 100%) was noted. AAV transduction of LTC-ICs was stable, with week-8 and -10 LTC-ICs showing comparable or better transduction relative to week-5 LTC-ICs. Fluorescence in situ hybridization (FISH) analyses performed to determine the fate of AAV vectors in transduced cells showed that 9% to 28% of CD34(+) and CD34(+)38(-) cells showed stable vector integration as evidenced by chromosome-associated signals in metaphase spreads. Comparisons of interphase and metaphase FISH suggested that a fraction of cells also contained episomal vector at early time points after transduction. Despite the apparent loss of the episomal forms with continued culture, the number of metaphases containing integrated vector genomes remained stable long term. Transgene transcription and placental alkaline phosphatase (PLAP) expression was observed in CD34(+), CD34(+)38(-) LTC-ICs in the absence of selective pressure. These results suggest that primitive myeloid progenitors are amenable to genetic modification with AAV vectors.
- Published
- 1999
39. Results of high-dose therapy and autologous bone marrow/stem cell transplantation during remission in poor-risk intermediate- and high-grade lymphoma: international index high and high-intermediate risk group.
- Author
-
Nademanee A, Molina A, O'Donnell MR, Dagis A, Snyder DS, Parker P, Stein A, Smith E, Planás I, Kashyap A, Spielberger R, Fung H, Wong KK, Somlo G, Margolin K, Chow W, Sniecinski I, Vora N, Blume KG, Niland J, and Forman SJ
- Subjects
- Adolescent, Adult, Combined Modality Therapy, Female, Humans, Lymphoma, Non-Hodgkin physiopathology, Male, Middle Aged, Pilot Projects, Prognosis, Remission Induction, Transplantation, Autologous, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Bone Marrow Purging, Hematopoietic Stem Cell Transplantation, Lymphoma, Non-Hodgkin therapy
- Abstract
We have conducted a pilot study to investigate the role of high-dose therapy and autologous bone marrow/stem cell transplantation (ASCT) during first complete or partial remission in 52 patients with poor-risk aggressive lymphoma. There were 42 patients with intermediate-grade or immunoblastic lymphoma who were considered to be high (60%) and high-intermediate risk (40%) groups at diagnosis based on the age-adjusted International Prognostic Index (IPI) and 10 patients with high-grade, SNCCL (small non-cleaved cell, Burkitt's, and non-Burkitt's), who at presentation had poor-risk features defined as elevated serum lactate dehydrogenase level, stage IV, and bulky mass >/=10 cm. The median age was 34 years (range, 16 to 56 years). Thirty-nine were transplanted in first complete remission and 13 in first partial remission after conventional therapy. Conditioning regimens consisted of total body irradiation (TBI) administered as a single fraction 750 cGy in 3 patients and in fractionated doses for a total of 1,200 cGy in 44 patients, in combination with 60 mg/kg etoposide and 100 mg/kg cyclophosphamide. Five patients with prior radiotherapy received 450 mg/m2 carmustine instead of TBI. Stem cell sources were either bone marrow and/or peripheral blood. No in vitro purging was used. All patients engrafted. Two SNCCL patients died of venoocclusive disease at 25 days and acute leukemia at 27 months posttransplantation. There were six relapses at 1.5 to 12.8 months posttransplantation. At a median follow-up of 44 months (range, 1 to 113 months), the estimated 3-year overall survival (OS) and disease-free survival (DFS) for all patients was 84% (95% confidence interval [CI], 70% to 92%) and 82% (95% CI, 68% to 91%), respectively. In the subset of patients with intermediate-grade and immunoblastic lymphoma, the 3-year DFS was 89% (95% CI, 74% to 96%) for all patients, 87% (95% CI, 67% to 96%) for high-risk patients, and 92% (95 CI, 61% to 99%) for high-intermediate risk patients. The 3-year OS and DFS for SNCCL patients were identical at 60% (95% CI, 30% to 84%). These results suggest that high-dose therapy and ASCT during first remission may improve the survival and prognosis of patients with poor-risk intermediate- and high-grade lymphoma. A prospective randomized study comparing high-dose therapy and ASCT with conventional chemotherapy in IPI high-risk patients with aggressive non-Hodgkin's lymphoma should be undertaken.
- Published
- 1997
40. Development of a candidate HLA A*0201 restricted peptide-based vaccine against human cytomegalovirus infection.
- Author
-
Diamond DJ, York J, Sun JY, Wright CL, and Forman SJ
- Subjects
- Animals, Cytomegalovirus Infections prevention & control, Cytotoxicity, Immunologic, Humans, Mice, Peptides immunology, Vaccines, Synthetic, Antigens, Viral immunology, Cytomegalovirus immunology, Cytomegalovirus Infections immunology, HLA-A Antigens immunology, T-Lymphocytes immunology, Viral Vaccines
- Abstract
The development of a protective cellular immune response against human cytomegalovirus (HCMV) is the most important determinant of recovery from HCMV infection after allogeneic bone marrow transplantation (BMT). The ultimate aim of our study is to develop an antigen-specific and peptide-based vaccine strategy against HCMV in the setting of BMT. Toward this end we have studied the cellular immune response against the immunodominant matrix protein pp65 of HCMV. Using an HLA A*0201-restricted T-cell clone reactive against pp65 from peripheral blood from a seropositive individual, we have mapped the position of the cytolytic T lymphocyte (CTL) epitope from HCMV pp65 to an 84-amino acid segment. Of the four peptides which best fit the HLA A*0201 motif in that region, one nonamer sensitized an autologous Epstein-Barr virus immortalized lymphocyte cell line for lysis. In vitro immunization of PBMC from HLA A*0201 and HCMV seropositive volunteers using the defined nonamer peptide stimulated significant recognition of HCMV infected or peptide-sensitized fibroblasts. Similarly, HLA A*0201 transgenic mice immunized with the nonamer peptide developed CTL that recognize both the immunizing peptide and endogenously processed pp65 in an HLA A*0201 restricted manner. Lipid modification of the amino terminus of the nonamer peptide resulted in its ability to stimulate immune responses without the use of adjuvant. This demonstration of a vaccine function of the nonamer peptide without adjuvant suggests its potential for use in an immunization trial of BMT donors to induce protective CTLs in patients undergoing allogeneic BMT.
- Published
- 1997
41. Hematopoietic potential and retroviral transduction of CD34+ Thy-1+ peripheral blood stem cells from asymptomatic human immunodeficiency virus type-1-infected individuals mobilized with granulocyte colony-stimulating factor.
- Author
-
Junker U, Moon JJ, Kalfoglou CS, Sniecinski I, Forman SJ, Zaia JA, Kaneshima H, and Böhnlein E
- Subjects
- Adult, Antigens, CD34 analysis, Blood Cell Count, Cell Separation, DNA, Viral blood, Feasibility Studies, Female, Flow Cytometry, Genetic Vectors, HIV Core Protein p24 blood, Hematopoietic Stem Cell Transplantation, Hematopoietic Stem Cells virology, Humans, Leukapheresis, Male, Middle Aged, Polymerase Chain Reaction, Thy-1 Antigens analysis, Viremia blood, Granulocyte Colony-Stimulating Factor pharmacology, HIV Infections blood, Hematopoiesis drug effects, Hematopoietic Stem Cells drug effects, Retroviridae genetics
- Abstract
The potential of hematopoietic stem cells (HSCs) from human immunodeficiency virus type-1 (HIV-1)-infected individuals, eg, self-renewal and multilineage differentiative capacity, might be perturbed due to the underlying disease. In this study, we assessed the HSC activity in the CD34+ Thy-1+ cell population of peripheral blood stem cells (PBSCs) of three asymptomatic HIV-1-infected individuals after granulocyte colony-stimulating factor (G-CSF; 10 microg/kg/d) mobilization. On day 4 of G-CSF treatment, 0.8% to 1% of the total blood mononuclear cells were CD34+. Leukapheresis followed by a two-step cell isolation process yielded a CD34+ Thy-1+ cell population of high purity (76% to 92% CD34+ Thy-1+ cells). This cell population showed no evidence of HIV-1-containing cells based on a semiquantitative HIV-1 DNA polymerase chain reaction. Furthermore, the purified cells showed normal hematopoietic potential in in vitro clonogenic assays. Successful gene transfer into committed progenitor cells (colony-forming units-cells) and more primitive stem/progenitor cells (long-term culture colony-forming cells) could be shown after amphotropic retroviral transduction. These data provide evidence that the CD34+ Thy-1+ stem cell compartment can be mobilized and enriched in early stage HIV-1-infected patients. Furthermore, successful transduction of this cell population as a prerequisite for stem cell-based clinical gene therapy protocols was demonstrated.
- Published
- 1997
42. Effect of CD34+ selection and various schedules of stem cell reinfusion and granulocyte colony-stimulating factor priming on hematopoietic recovery after high-dose chemotherapy for breast cancer.
- Author
-
Somlo G, Sniecinski I, Odom-Maryon T, Nowicki B, Chow W, Hamasaki V, Leong L, Margolin K, Morgan R Jr, Raschko J, Shibata S, Tetef M, Molina A, Berenson RJ, Forman SJ, and Doroshow JH
- Subjects
- Adult, Antigens, CD34, Blood Cell Count, Cell Separation, Cisplatin administration & dosage, Combined Modality Therapy, Cyclophosphamide administration & dosage, Etoposide administration & dosage, Female, Hematopoietic Stem Cells pathology, Humans, Middle Aged, Antineoplastic Combined Chemotherapy Protocols administration & dosage, Breast Neoplasms therapy, Carcinoma therapy, Granulocyte Colony-Stimulating Factor administration & dosage, Hematopoietic Stem Cell Transplantation, Hematopoietic Stem Cells drug effects
- Abstract
We evaluated the effects of various schedules of peripheral blood stem cell (PBSC) reinfusion, granulocyte colony-stimulating factor (G-CSF) priming, and CD34+ enrichment on hematopoietic recovery in 88 patients with advanced breast cancer treated with high-dose chemotherapy, consisting of cisplatin 250 mg/m2, etoposide 60 mg/kg, and cyclophosphamide 100 mg/kg. PBSC (> or = 7.5 x 10(8) nucleated cells/kg) were collected following priming with G-CSF and were either immediately cryopreserved (48 patients; cohorts A and B) or were first processed for CD34+ enrichment (40 patients; cohorts C and D). Patients in cohorts A and C received PBSC on day 0; patients in cohorts B and D received 25% of their nucleated cells on day -2 and 75% on day 0 (split reinfusion). Patients in cohorts A, B, and C were primed with G-CSF 10 micrograms/kg, subcutaneously (SC), once a day; patients in cohort D were primed with 5 micrograms/kg G-CSF, SC, twice daily (bid). Bid administration of G-CSF yielded 2.3 to 4.7 x higher numbers of CD34+ cells in the PBSC product than the same total dose given once a day (P = .002). Reinfusion of 25% of unselected PBSC on day -2 (median, 2.26 x 10(8)/kg nucleated cells [range, 1.7 to 3.3 x 10(8)/kg]) with the remaining cells reinfused on day 0 resulted in earlier granulocyte recovery to > or = 500/microL when compared with reinfusion of all stem cells on day 0 (group B, median of 8 days [range, 7 to 11] v group A, 10 days [range, 8 to 11], P = .0003); no schedule-dependent difference was noted in reaching platelet independence (group B, 11.5 days [range, 5 to 21]; group A, 12 days [range, 8 to 24], P = not significant). Split schedule reinfusion of CD34(+)-selected PBSC did not accelerate granulocyte recovery. In groups D and C, the median number of days to granulocyte recovery was 12 (range, 8 to 22) and 11.5 (range, 9 to 13); patients became platelet independent by day 15 (range, 6 to 22) and 14 (range, 12 to 23), respectively. CD34(+)-selected PBSC rescue decreased the incidence of postreinfusion nausea, emesis, and oxygen desaturation in comparison to unselected PBSC reinfusion (P < or = .005 for each). Hematopoietic recovery may be accelerated by earlier reinfusion of approximately 2.26 x 10(8)/kg unselected nucleated cells. Earlier recovery may be triggered by components other than the progenitors included in the CD34+ cell population. Sustained hematopoietic recovery can also be achieved with CD34(+)-selected PBSC alone. Dosing of G-CSF on a bid schedule generates higher CD34+ cell yield in the leukapheresis product. Whether even earlier "sacrificial" reinfusion of approximately 2 x 10(8)/kg unselected nucleated cells concomitant with the administration of high-dose chemotherapy would reduce the duration of absolute granulocytopenia further while initiating sustained long-term hematopoietic recovery will require further investigation.
- Published
- 1997
43. Integration of adeno-associated virus vectors in CD34+ human hematopoietic progenitor cells after transduction.
- Author
-
Fisher-Adams G, Wong KK Jr, Podsakoff G, Forman SJ, and Chatterjee S
- Subjects
- Amino Acid Sequence, Antigens, CD34 analysis, Bone Marrow Cells, Cell Cycle, Chromosome Mapping, DNA Replication, Fetal Blood cytology, Hematopoietic Stem Cells metabolism, Humans, In Situ Hybridization, Fluorescence, Molecular Sequence Data, Recombinant Fusion Proteins biosynthesis, Transfection, Dependovirus genetics, Genetic Vectors genetics, Hematopoietic Stem Cells virology, Virus Integration
- Abstract
Gene transfer vectors based on adeno-associated virus (AAV) appear promising because of their high transduction frequencies regardless of cell cycle status and ability to integrate into chromosomal DNA. We tested AAV-mediated gene transfer into a panel of human bone marrow or umbilical cord-derived CD34+ hematopoietic progenitor cells, using vectors encoding several transgenes under the control of viral and cellular promoters. Gene transfer was evaluated by (1) chromosomal integration of vector sequences and (2) analysis of transgene expression. Southern hybridization and fluorescence in situ hybridization analysis of transduced CD34 genomic DNA showed the presence of integrated vector sequences in chromosomal DNA in a portion of transduced cells and showed that integrated vector sequences were replicated along with cellular DNA during mitosis. Transgene expression in transduced CD34 cells in suspension cultures and in myeloid colonies differentiating in vitro from transduced CD34 cells approximated that predicted by the multiplicity of transduction. This was true in CD34 cells from different donors, regardless of the transgene or selective pressure. Comparisons of CD34 cell transduction either before or after cytokine stimulation showed similar gene transfer frequencies. Our findings suggest that AAV transduction of CD34+ hematopoietic progenitor cells is efficient, can lead to stable integration in a population of transduced cells, and may therefore provide the basis for safe and efficient ex vivo gene therapy of the hematopoietic system.
- Published
- 1996
44. Molecular analysis of T-cell receptor repertoire in bone marrow transplant recipients: evidence for oligoclonal T-cell expansion in graft-versus-host disease lesions.
- Author
-
Liu X, Chesnokova V, Forman SJ, and Diamond DJ
- Subjects
- Adolescent, Adult, Base Sequence, Bone Marrow Transplantation adverse effects, Clone Cells, Humans, Middle Aged, Molecular Sequence Data, Polymerase Chain Reaction, Receptors, Antigen, T-Cell immunology, Sequence Analysis, Bone Marrow Transplantation immunology, Graft vs Host Disease immunology, Receptors, Antigen, T-Cell genetics
- Abstract
We have analyzed the T-cell receptor (TCR) V beta repertoire using polymerase chain reaction (PCR) in a cohort of eight patients receiving allogeneic bone marrow transplantation (BMT) from related and unrelated donors at the City of Hope. Results of PCR studies from graft-versus-host disease (GVHD) skin lesions show a bias in the usage of TCR V beta families, whereas examination of peripheral blood (PB) withdrawn at the same time did not reveal a similar phenomenon. In one such family, TCR V beta 2 is predominantly expressed in 7 of 7 biopsy specimens examined. V beta 2 TCR expression from these patients was analyzed more extensively using a combination of individual TCR gene cloning, followed by sequence analysis. We found evidence of oligoclonal expansion of single V beta 2-bearing TCRs in GVHD lesions, and in the PB of some patients after diagnosis of GVHD. In contrast, GVHD-negative biopsy samples showed no evidence for clonotypic TCR amplification. Sequence-specific TCR CDR3 region probes were derived from analysis of the predominant expressed TCR in GVHD lesions, and used to probe Southern blots of amplified V beta 2 TCR mRNA from PB and tissue from BMT recipients and their respective donors. In most cases the probes are highly specific in detecting TCR expression from GVHD lesions alone, although in several instances expression could be detected in PB after GVHD diagnosis. These data provide supporting evidence for the hypothesis that acute GVHD is associated with expansion of T-cell clones expressing antigen-specific TCRs that may contribute to the disease pathology.
- Published
- 1996
45. Thalidomide as salvage therapy for chronic graft-versus-host disease.
- Author
-
Parker PM, Chao N, Nademanee A, O'Donnell MR, Schmidt GM, Snyder DS, Stein AS, Smith EP, Molina A, Stepan DE, Kashyap A, Planas I, Spielberger R, Somlo G, Margolin K, Zwingenberger K, Wilsman K, Negrin RS, Long GD, Niland JC, Blume KG, and Forman SJ
- Subjects
- Adolescent, Adult, Bone Marrow Transplantation mortality, Child, Chronic Disease, Constipation chemically induced, Cyclosporine therapeutic use, Female, Graft vs Host Disease mortality, Hematologic Diseases therapy, Humans, Infections mortality, Leukemia therapy, Male, Middle Aged, Neuritis chemically induced, Prednisone therapeutic use, Remission Induction, Survival Rate, Thalidomide adverse effects, Treatment Outcome, Bone Marrow Transplantation adverse effects, Drug Eruptions etiology, Graft vs Host Disease drug therapy, Immunosuppressive Agents therapeutic use, Neutropenia chemically induced, Salvage Therapy, Thalidomide therapeutic use
- Abstract
Thalidomide has been reported to be an effective agent for treatment of chronic graft-versus-host disease (CGVHD). To determine the efficacy of this agent in patients with refractory CGVHD a total of 80 patients who failed to respond to prednisone (PSE) or PSE and cyclosporine (CSA) were treated with thalidomide. Sixteen patients (20%) had a sustained response, 9 with a complete remission and 7 with a partial response. Twenty-nine patients (36%) had thalidomide discontinued because of side effects, which included sedation, constipation, neuritis, skin rash, and neutropenia. Side effects were reversible with drug discontinuation except for mild residual neuritis in one case. Rashes and neutropenia have not previously been reported as thalidomide side effects when used for CGVHD treatment. We conclude thalidomide is immunosuppressive and active in the treatment of CGVHD. A high incidence of reversible side effects limited dose intensity and reduced the number of patients who could benefit from treatment.
- Published
- 1995
46. Long-term follow-up of allogeneic bone marrow recipients for Philadelphia chromosome-positive acute lymphoblastic leukemia.
- Author
-
Chao NJ, Blume KG, Forman SJ, and Snyder DS
- Subjects
- Disease-Free Survival, Follow-Up Studies, Humans, Leukemia, Myelogenous, Chronic, BCR-ABL Positive mortality, Remission Induction, Retrospective Studies, Treatment Outcome, Bone Marrow Transplantation, Leukemia, Myelogenous, Chronic, BCR-ABL Positive therapy
- Published
- 1995
47. Clonal karyotypic hematopoietic cell abnormalities occurring after autologous bone marrow transplantation for Hodgkin's disease and non-Hodgkin's lymphoma.
- Author
-
Traweek ST, Slovak ML, Nademanee AP, Brynes RK, Niland JC, and Forman SJ
- Subjects
- Adult, Chromosome Disorders, Clone Cells, Combined Modality Therapy, Female, Hematopoietic Stem Cells pathology, Hodgkin Disease drug therapy, Hodgkin Disease surgery, Humans, Karyotyping, Lymphoma, Non-Hodgkin drug therapy, Lymphoma, Non-Hodgkin surgery, Male, Time Factors, Transplantation, Autologous, Bone Marrow Transplantation pathology, Chromosome Aberrations etiology, Hodgkin Disease therapy, Lymphoma, Non-Hodgkin therapy, Myelodysplastic Syndromes etiology, Neoplasms, Second Primary etiology
- Abstract
Over a 6-year period, 275 patients were treated with autologous bone marrow transplantation (auto-BMT) for advanced-stage malignant lymphoma. After BMT, clonal chromosomal abnormalities were detected in hematopoietic cells from 10 patients. All 10 had morphologically and cytogenetically normal BMs at the time of stem cell harvest. The cytogenetic changes were first detected 1.8 to 6.5 years (mean, 3.9) after induction chemotherapy, and 0.5 to 3.1 years (mean, 1.4) after transplantation, and were characteristic of those reported for therapy-related myelodysplastic syndrome (MDS) in 9 of the patients: abnormalities of chromosome 5 or 7 (classical-form) were present in 4, 11q23 or 21q22 abnormalities (topoisomerase II-related form) were detected in 3, and a combination of both forms was seen in 2 patients. Clonal 2p abnormalities were found in the 1 remaining patient. The abnormal karyotypes were associated with morphologically recognizable MDS in 3 patients and with acute myeloid leukemia (AML) arising in MDS in 2. Four of these patients have died: 3 of AML and 1 of infection. One patient is still alive with cytopenia. The clonal cytogenetic abnormalities were not associated with MDS in 5 patients: 1 has died of recurrent lymphoma, 2 have cytopenia, and 2 still have no morphologic or clinical evidence of MDS after short follow-up (4 and 13 months). Compared with a control group matched for disease, length of follow-up, and treatment with auto-BMT, there were no statistically significant associations between the development of clonal chromosomal abnormalities and age, number of chemotherapeutic regimens, prior local radiation, BMT conditioning regimen (with or without total body irradiation), or type of lymphoma. These studies show that the risk of developing clonal cytogenetic changes after auto-BMT for malignant lymphoma is approximately 9% at 3 years, even when pre-BMT karyotypic studies are normal. The exact significance of these cytogenetic abnormalities in the absence of MDS or AML is unclear.
- Published
- 1994
48. Treatment and prevention of cytomegalovirus pneumonia after bone marrow transplantation: where do we stand?
- Author
-
Forman SJ and Zaia JA
- Subjects
- Cytomegalovirus Infections diagnosis, Cytomegalovirus Infections etiology, Cytomegalovirus Infections immunology, Humans, Pneumonia, Viral diagnosis, Pneumonia, Viral etiology, Pneumonia, Viral immunology, Risk Factors, Bone Marrow Transplantation, Cytomegalovirus Infections drug therapy, Cytomegalovirus Infections prevention & control, Pneumonia, Viral drug therapy, Pneumonia, Viral prevention & control
- Published
- 1994
49. Ribozyme-mediated inhibition of bcr-abl gene expression in a Philadelphia chromosome-positive cell line.
- Author
-
Snyder DS, Wu Y, Wang JL, Rossi JJ, Swiderski P, Kaplan BE, and Forman SJ
- Subjects
- Base Sequence, Blast Crisis, Cell Division, Humans, Leukemia, Myelogenous, Chronic, BCR-ABL Positive pathology, Magnesium pharmacology, Molecular Sequence Data, RNA, Catalytic genetics, RNA, Messenger chemistry, Ribonucleases metabolism, Transfection, Tumor Cells, Cultured, Fusion Proteins, bcr-abl genetics, Gene Expression, Leukemia, Myelogenous, Chronic, BCR-ABL Positive genetics, RNA, Catalytic metabolism, RNA, Messenger metabolism
- Abstract
The bcr-abl fusion gene is the molecular counterpart of the Philadelphia chromosome (Ph1) and is directly involved in the pathogenesis of Ph1+ leukemia. Inhibition of bcr-abl gene expression may have profound effects on the cell biology of Ph1+ cells, as recent experiments with antisense oligonucleotides have shown. In this study we have designed and synthesized a unique ribozyme that is directed against bcr-abl mRNA. The ribozyme cleaved bcr-abl mRNA in a cell-free in vitro system. A DNA-RNA hybrid ribozyme was then incorporated into a liposome vector and transfected into EM-2 cells, a cell line derived from a patient with blast crisis of chronic myelogenous leukemia. The ribozyme decreased levels of detectable bcr-abl mRNA in these cells, inhibited expression of the bcr-abl gene product, p210bcr-abl, and inhibited cell growth. This anti-bcr-abl ribozyme may be a useful tool to study the cell biology of Ph1+ leukemia and may ultimately have therapeutic potential in treating patients with Ph1 leukemias.
- Published
- 1993
50. A prospective randomized comparison of total body irradiation-etoposide versus busulfan-cyclophosphamide as preparatory regimens for bone marrow transplantation in patients with leukemia who were not in first remission: a Southwest Oncology Group study.
- Author
-
Blume KG, Kopecky KJ, Henslee-Downey JP, Forman SJ, Stiff PJ, LeMaistre CF, and Appelbaum FR
- Subjects
- Adolescent, Adult, Aged, Busulfan administration & dosage, Child, Child, Preschool, Combined Modality Therapy, Cyclophosphamide administration & dosage, Etoposide administration & dosage, Female, Humans, Male, Middle Aged, Prospective Studies, Remission Induction, Risk Factors, Bone Marrow Transplantation, Busulfan therapeutic use, Cyclophosphamide therapeutic use, Etoposide therapeutic use, Leukemia therapy, Whole-Body Irradiation
- Abstract
Two novel preparatory regimens for conditioning of patients with leukemia for allogeneic bone marrow transplantation (BMT) from histocompatible sibling donors have been tested in a phase III trial under the auspices of the Southwest Oncology Group (SWOG 8612). These two regimens consisted either of fractionated total body irradiation and etoposide (FTBI/VP-16) or high-dose busulfan with cyclophosphamide (BU/CY). Only patients who had failed prior conventional management at least once were study eligible, ie, no patients with acute leukemia in first remission (CR) or in first chronic phase (CP) of chronic myelogenous leukemia (CML) participated. Patients were stratified according to the following risk criteria: "good-risk" patients were those who were in second CR of their acute leukemia or in accelerated phase (AP) of CML; "poor-risk" patients had further advanced stages of leukemia. During a 52-month period, 131 patients were registered of whom 122 (93%) were study eligible. Sixty-one eligible patients were randomized to the FTBI/VP-16 arm and 61 to the BU/CY regimen. Of these 122 patients, 114 (93%) proceeded to BMT according to protocol. Posttransplant immunosuppression to prevent graft-versus-host disease (GVHD) consisted of cyclosporine and prednisone (CSA/PSE). Neither overall survival nor disease-free survival (DFS) differed significantly between the two treatment groups (P = .89 and .69, respectively). Estimated DFS for "good-risk" patients who had been prepared with the FTBI/VP-16 regimen was 55% +/- 11%, as compared with patients treated with BU/CY whose DFS figure was 34% +/- 10% (P = .30). For "poor-risk" candidates, the DFS rates at 24 months were 17% +/- 6% (for FTBI/VP-16) and 24% +/- 8% (for BU/CY), respectively (P = .81). These figures do not differ significantly, especially in view of the fact that the "good-risk" patients prepared with the FTBI/VP-16 regimen were younger than those treated with BU/CY. Both regimens were well tolerated with no regimen-related deaths encountered during the 6-week period after BMT. This study also confirmed the efficacy of the CSA/PSE combination in the prevention of GVHD with 23 of 113 (20%) of BMT recipients developing moderate to severe acute GVHD. The leading cause for treatment failure was leukemic relapse (45 of the 114 BMT recipients suffered a recurrence of their leukemia), whereas 38 patients died without evidence of relapse. Thirty-one patients are alive and in continued CR after marrow transplantation; four are alive in relapse.(ABSTRACT TRUNCATED AT 400 WORDS)
- Published
- 1993
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.