Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Ted J. Hofmann is active.

Publication


Featured researches published by Ted J. Hofmann.


Proceedings of the National Academy of Sciences of the United States of America | 2002

Isolated allogeneic bone marrow-derived mesenchymal cells engraft and stimulate growth in children with osteogenesis imperfecta: Implications for cell therapy of bone

Edwin M. Horwitz; Patricia L. Gordon; Winston K. K. Koo; Jeffrey C. Marx; Michael D. Neel; Rene Y. McNall; Linda M. Muul; Ted J. Hofmann

Treatment with isolated allogeneic mesenchymal cells has the potential to enhance the therapeutic effects of conventional bone marrow transplantation in patients with genetic disorders affecting mesenchymal tissues, including bone, cartilage, and muscle. To demonstrate the feasibility of mesenchymal cell therapy and to gain insight into the transplant biology of these cells, we used gene-marked, donor marrow-derived mesenchymal cells to treat six children who had undergone standard bone marrow transplantation for severe osteogenesis imperfecta. Each child received two infusions of the allogeneic cells. Five of six patients showed engraftment in one or more sites, including bone, skin, and marrow stroma, and had an acceleration of growth velocity during the first 6 mo postinfusion. This improvement ranged from 60% to 94% (median, 70%) of the predicted median values for age- and sex-matched unaffected children, compared with 0% to 40% (median, 20%) over the 6 mo immediately preceding the infusions. There was no clinically significant toxicity except for an urticarial rash in one patient just after the second infusion. Failure to detect engraftment of cells expressing the neomycin phosphotransferase marker gene suggested the potential for immune attack against therapeutic cells expressing a foreign protein. Thus, allogeneic mesenchymal cells offer feasible posttransplantation therapy for osteogenesis imperfecta and likely other disorders originating in mesenchymal precursors.


Cancer Discovery | 2015

Convergence of Acquired Mutations and Alternative Splicing of CD19 Enables Resistance to CART-19 Immunotherapy

Elena Sotillo; David M. Barrett; Kathryn L. Black; Asen Bagashev; Derek A. Oldridge; Glendon Wu; Robyn T. Sussman; Claudia Lanauze; Marco Ruella; Matthew R. Gazzara; Nicole M. Martinez; Colleen T. Harrington; Elaine Y. Chung; Jessica Perazzelli; Ted J. Hofmann; Shannon L. Maude; Pichai Raman; Alejandro Barrera; Saar Gill; Simon F. Lacey; J. Joseph Melenhorst; David Allman; Elad Jacoby; Terry J. Fry; Crystal L. Mackall; Yoseph Barash; Kristen W. Lynch; John M. Maris; Stephan A. Grupp; Andrei Thomas-Tikhonenko

UNLABELLED The CD19 antigen, expressed on most B-cell acute lymphoblastic leukemias (B-ALL), can be targeted with chimeric antigen receptor-armed T cells (CART-19), but relapses with epitope loss occur in 10% to 20% of pediatric responders. We detected hemizygous deletions spanning the CD19 locus and de novo frameshift and missense mutations in exon 2 of CD19 in some relapse samples. However, we also discovered alternatively spliced CD19 mRNA species, including one lacking exon 2. Pull-down/siRNA experiments identified SRSF3 as a splicing factor involved in exon 2 retention, and its levels were lower in relapsed B-ALL. Using genome editing, we demonstrated that exon 2 skipping bypasses exon 2 mutations in B-ALL cells and allows expression of the N-terminally truncated CD19 variant, which fails to trigger killing by CART-19 but partly rescues defects associated with CD19 loss. Thus, this mechanism of resistance is based on a combination of deleterious mutations and ensuing selection for alternatively spliced RNA isoforms. SIGNIFICANCE CART-19 yield 70% response rates in patients with B-ALL, but also produce escape variants. We discovered that the underlying mechanism is the selection for preexisting alternatively spliced CD19 isoforms with the compromised CART-19 epitope. This mechanism suggests a possibility of targeting alternative CD19 ectodomains, which could improve survival of patients with B-cell neoplasms.


Blood | 2009

Restoration and reversible expansion of the osteoblastic hematopoietic stem cell niche after marrow radioablation

Massimo Dominici; Valeria Rasini; Rita Bussolari; Xiaohua Chen; Ted J. Hofmann; Carlotta Spano; Daniela Bernabei; Elena Veronesi; Filippo Bertoni; Paolo Paolucci; Pierfranco Conte; Edwin M. Horwitz

Adequate recovery of hematopoietic stem cell (HSC) niches after cytotoxic conditioning regimens is essential to successful bone marrow transplantation. Yet, very little is known about the mechanisms that drive the restoration of these niches after bone marrow injury. Here we describe a profound disruption of the marrow microenvironment after lethal total body irradiation of mice that leads to the generation of osteoblasts restoring the HSC niche, followed by a transient, reversible expansion of this niche. Within 48 hours after irradiation, surviving host megakaryocytes were observed close to the endosteal surface of trabecular bone rather than in their normal parasinusoidal site concomitant with an increased stromal-derived factor-1 level. A subsequent increase in 2 megakaryocyte-derived growth factors, platelet-derived growth factor-beta and basic fibroblast growth factor, induces a 2-fold expansion of the population of N-cadherin-/osteopontin-positive osteoblasts, relative to the homeostatic osteoblast population, and hence, increases the number of potential niches for HSC engraftment. After donor cell engraftment, this expanded microenvironment reverts to its homeostatic state. Our results demonstrate the rapid recovery of osteoblastic stem cell niches after marrow radioablation, provide critical insights into the associated mechanisms, and suggest novel means to manipulate the bone marrow microenvironment to promote HSC engraftment.


Blood | 2012

Transplanted bone marrow mononuclear cells and MSCs impart clinical benefit to children with osteogenesis imperfecta through different mechanisms

Satoru Otsuru; Patricia L. Gordon; Kengo Shimono; Reena Jethva; Roberta Marino; Charlotte L. Phillips; Ted J. Hofmann; Elena Veronesi; Massimo Dominici; Masahiro Iwamoto; Edwin M. Horwitz

Transplantation of whole bone marrow (BMT) as well as ex vivo-expanded mesenchymal stromal cells (MSCs) leads to striking clinical benefits in children with osteogenesis imperfecta (OI); however, the underlying mechanism of these cell therapies has not been elucidated. Here, we show that non-(plastic)-adherent bone marrow cells (NABMCs) are more potent osteoprogenitors than MSCs in mice. Translating these findings to the clinic, a T cell-depleted marrow mononuclear cell boost (> 99.99% NABMC) given to children with OI who had previously undergone BMT resulted in marked growth acceleration in a subset of patients, unambiguously indicating the therapeutic potential of bone marrow cells for these patients. Then, in a murine model of OI, we demonstrated that as the donor NABMCs differentiate to osteoblasts, they contribute normal collagen to the bone matrix. In contrast, MSCs do not substantially engraft in bone, but secrete a soluble mediator that indirectly stimulates growth, data which provide the underlying mechanism of our prior clinical trial of MSC therapy for children with OI. Collectively, our data indicate that both NABMCs and MSCs constitute effective cell therapy for OI, but exert their clinical impact by different, complementary mechanisms. The study is registered at www.clinicaltrials.gov as NCT00187018.


Journal of Clinical Investigation | 2016

Dual CD19 and CD123 targeting prevents antigen-loss relapses after CD19-directed immunotherapies

Marco Ruella; David M. Barrett; Saad S. Kenderian; Olga Shestova; Ted J. Hofmann; Jessica Perazzelli; Michael Klichinsky; Vania Aikawa; Farzana Nazimuddin; Miroslaw Kozlowski; John Scholler; Simon F. Lacey; J. Joseph Melenhorst; Jennifer J.D. Morrissette; David A. Christian; Christopher A. Hunter; Michael Kalos; David L. Porter; Carl H. June; Stephan A. Grupp; Saar Gill

Potent CD19-directed immunotherapies, such as chimeric antigen receptor T cells (CART) and blinatumomab, have drastically changed the outcome of patients with relapsed/refractory B cell acute lymphoblastic leukemia (B-ALL). However, CD19-negative relapses have emerged as a major problem that is observed in approximately 30% of treated patients. Developing approaches to preventing and treating antigen-loss escapes would therefore represent a vertical advance in the field. Here, we found that in primary patient samples, the IL-3 receptor α chain CD123 was highly expressed on leukemia-initiating cells and CD19-negative blasts in bulk B-ALL at baseline and at relapse after CART19 administration. Using intravital imaging in an antigen-loss CD19-negative relapse xenograft model, we determined that CART123, but not CART19, recognized leukemic blasts, established protracted synapses, and eradicated CD19-negative leukemia, leading to prolonged survival. Furthermore, combining CART19 and CART123 prevented antigen-loss relapses in xenograft models. Finally, we devised a dual CAR-expressing construct that combined CD19- and CD123-mediated T cell activation and demonstrated that it provides superior in vivo activity against B-ALL compared with single-expressing CART or pooled combination CART. In conclusion, these findings indicate that targeting CD19 and CD123 on leukemic blasts represents an effective strategy for treating and preventing antigen-loss relapses occurring after CD19-directed therapies.


Blood | 2013

Megakaryocytes promote murine osteoblastic HSC niche expansion and stem cell engraftment after radioablative conditioning

Timothy S. Olson; Anna Caselli; Satoru Otsuru; Ted J. Hofmann; Richard T. Williams; Paolo Paolucci; Massimo Dominici; Edwin M. Horwitz

Successful hematopoietic stem cell (HSC) transplantation requires donor HSC engraftment within specialized bone marrow microenvironments known as HSC niches. We have previously reported a profound remodeling of the endosteal osteoblastic HSC niche after total body irradiation (TBI), defined as relocalization of surviving megakaryocytes to the niche site and marked expansion of endosteal osteoblasts. We now demonstrate that host megakaryocytes function critically in expansion of the endosteal niche after preparative radioablation and in the engraftment of donor HSC. We show that TBI-induced migration of megakaryocytes to the endosteal niche depends on thrombopoietin signaling through the c-MPL receptor on megakaryocytes, as well as CD41 integrin-mediated adhesion. Moreover, niche osteoblast proliferation post-TBI required megakaryocyte-secreted platelet-derived growth factor-BB. Furthermore, blockade of c-MPL-dependent megakaryocyte migration and function after TBI resulted in a significant decrease in donor HSC engraftment in primary and competitive secondary transplantation assays. Finally, we administered thrombopoietin to mice beginning 5 days before marrow radioablation and ending 24 hours before transplant to enhance megakaryocyte function post-TBI, and found that this strategy significantly enhanced donor HSC engraftment, providing a rationale for improving hematopoietic recovery and perhaps overall outcome after clinical HSC transplantation.


Blood | 2008

Donor cell–derived osteopoiesis originates from a self-renewing stem cell with a limited regenerative contribution after transplantation

Massimo Dominici; Roberta Marino; Valeria Rasini; Carlotta Spano; Paolo Paolucci; Pierfranco Conte; Ted J. Hofmann; Edwin M. Horwitz

In principle, bone marrow transplantation should offer effective treatment for disorders originating from defects in mesenchymal stem cells. Results with the bone disease osteogenesis imperfecta support this hypothesis, although the rate of clinical improvement seen early after transplantation does not persist long term, raising questions as to the regenerative capacity of the donor-derived mesenchymal progenitors. We therefore studied the kinetics and histologic/anatomic pattern of osteopoietic engraftment after transplantation of GFP-expressing nonadherent marrow cells in mice. Serial tracking of donor-derived GFP(+) cells over 52 weeks showed abundant clusters of donor-derived osteoblasts/osteocytes in the epiphysis and metaphysis but not the diaphysis, a distribution that paralleled the sites of initial hematopoietic engraftment. Osteopoietic chimerism decreased from approximately 30% to 10% by 24 weeks after transplantation, declining to negligible levels thereafter. Secondary transplantation studies provided evidence for a self-renewing osteopoietic stem cell in the marrow graft. We conclude that a transplantable, primitive, self-renewing osteopoietic cell within the nonadherent marrow cell population engrafts in an endosteal niche, like hematopoietic stem cells, and regenerates a significant fraction of all bone cells. The lack of durable donor-derived osteopoiesis may reflect an intrinsic genetic program or exogenous environmental signaling that suppresses the differentiation capacity of the donor stem cells.


Stem Cells | 2014

Tendon Progenitor Cells in Injured Tendons Have Strong Chondrogenic Potential: The CD105-Negative Subpopulation Induces Chondrogenic Degeneration

Shuji Asai; Satoru Otsuru; Maria Elena Candela; Leslie Cantley; Kenta Uchibe; Ted J. Hofmann; Kairui Zhang; Keith L. Wapner; Louis J. Soslowsky; Edwin M. Horwitz; Motomi Enomoto-Iwamoto

To study the cellular mechanism of the tendon repair process, we used a mouse Achilles tendon injury model to focus on the cells recruited to the injured site. The cells isolated from injured tendon 1 week after the surgery and uninjured tendons contained the connective tissue progenitor populations as determined by colony‐forming capacity, cell surface markers, and multipotency. When the injured tendon‐derived progenitor cells (inTPCs) were transplanted into injured Achilles tendons, they were not only integrated in the regenerating area expressing tenogenic phenotype but also trans‐differentiated into chondrogenic cells in the degenerative lesion that underwent ectopic endochondral ossification. Surprisingly, the micromass culture of the inTPCs rapidly underwent chondrogenic differentiation even in the absence of exogenous bone morphogenetic proteins or TGFβs. The cells isolated from human ruptured tendon tissues also showed connective tissue progenitor properties and exhibited stronger chondrogenic ability than bone marrow stromal cells. The mouse inTPCs contained two subpopulations one positive and one negative for CD105, a coreceptor of the TGFβ superfamily. The CD105‐negative cells showed superior chondrogenic potential in vitro and induced larger chondroid degenerative lesions in mice as compared to the CD105‐positive cells. These findings indicate that tendon progenitor cells are recruited to the injured site of tendons and have a strong chondrogenic potential and that the CD105‐negative population of these cells would be the cause for chondroid degeneration in injured tendons. The newly identified cells recruited to the injured tendon may provide novel targets to develop therapeutic strategies to facilitate tendon repair. Stem Cells 2014;32:3266–3277


Cytotherapy | 2013

Improved isolation and expansion of bone marrow mesenchymal stromal cells using a novel marrow filter device

Satoru Otsuru; Ted J. Hofmann; Timothy S. Olson; Massimo Dominici; Edwin M. Horwitz

BACKGROUND AIMS Mesenchymal stromal cells (MSCs) have been studied as cell therapy to treat a vast array of diseases. In clinical MSC production, the isolated cells must undergo extensive ex vivo expansion to obtain a sufficient dose of MSCs for the investigational treatment. However, extended tissue culture is fraught with potential hazards, including contamination and malignant transformation. Changes of gene expression with prolonged culture may alter the therapeutic potential of the cells. Increasing the recovery of MSCs from the freshly harvested bone marrow allowing for less ex vivo expansion would represent a major advance in MSC therapy. METHODS Human bone marrow cells from eight healthy donors were processed using a marrow filter device and, in parallel, using buoyant density centrifugation by two independent investigators. The initial nucleated cell recovery and the final yield, immunophenotype and trilineage differentiation potential of second-passage MSCs were examined. RESULTS The marrow filter device generated significantly greater initial cell recovery requiring less investigator time and resulted in approximately 2.5-fold more MSCs after the second passage. The immunophenotype and differentiation potential of MSCs isolated using the two methods were equivalent and consistent with the defining criteria. The two independent investigators generated comparable results. CONCLUSIONS This novel filter device is a fast, efficient and reliable system to isolate MSCs and should greatly expedite pre-clinical and clinical investigations of MSC therapy.


Stem Cells | 2013

IGF‐1‐mediated osteoblastic niche expansion enhances long‐term hematopoietic stem cell engraftment after murine bone marrow transplantation

Anna Caselli; Timothy S. Olson; Satoru Otsuru; Xiaohua Chen; Ted J. Hofmann; Hyun-Duck Nah; Giulia Grisendi; Paolo Paolucci; Massimo Dominici; Edwin M. Horwitz

The efficiency of hematopoietic stem cell (HSC) engraftment after bone marrow (BM) transplantation depends largely on the capacity of the marrow microenvironment to accept the transplanted cells. While radioablation of BM damages osteoblastic stem cell niches, little is known about their restoration and mechanisms governing their receptivity to engraft transplanted HSCs. We previously reported rapid restoration and profound expansion of the marrow endosteal microenvironment in response to marrow radioablation. Here, we show that this reorganization represents proliferation of mature endosteal osteoblasts which seem to arise from a small subset of high‐proliferative, relatively radio‐resistant endosteal cells. Multiple layers of osteoblasts form along the endosteal surface within 48 hours after total body irradiation, concomitant with a peak in marrow cytokine expression. This niche reorganization fosters homing of the transplanted hematopoietic cells to the host marrow space and engraftment of long‐term‐HSC. Inhibition of insulin‐like growth factor (IGF)‐1‐receptor tyrosine kinase signaling abrogates endosteal osteoblast proliferation and donor HSC engraftment, suggesting that the cytokine IGF‐1 is a crucial mediator of endosteal niche reorganization and consequently donor HSC engraftment. Further understanding of this novel mechanism of IGF‐1‐dependent osteoblastic niche expansion and HSC engraftment may yield clinical applications for improving engraftment efficiency after clinical HSC transplantation. Stem Cells 2013;31:2193–2204

Collaboration


Dive into the Ted J. Hofmann's collaboration.

Top Co-Authors

Avatar

Edwin M. Horwitz

Nationwide Children's Hospital

View shared research outputs
Top Co-Authors

Avatar

Massimo Dominici

University of Modena and Reggio Emilia

View shared research outputs
Top Co-Authors

Avatar

Satoru Otsuru

The Research Institute at Nationwide Children's Hospital

View shared research outputs
Top Co-Authors

Avatar

Timothy S. Olson

Children's Hospital of Philadelphia

View shared research outputs
Top Co-Authors

Avatar

Valeria Rasini

University of Modena and Reggio Emilia

View shared research outputs
Top Co-Authors

Avatar

Roberta Marino

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar

Paolo Paolucci

University of Modena and Reggio Emilia

View shared research outputs
Top Co-Authors

Avatar

David M. Barrett

Children's Hospital of Philadelphia

View shared research outputs
Top Co-Authors

Avatar

Elena Veronesi

University of Modena and Reggio Emilia

View shared research outputs
Top Co-Authors

Avatar

Stephan A. Grupp

Children's Hospital of Philadelphia

View shared research outputs
Researchain Logo
Decentralizing Knowledge