Advertisement
100th Anniversary Viewpoints Open Access | 10.1172/JCI179944
1Division of Pediatric Hematology and Oncology, Boston Children’s Hospital and the Dana-Farber Cancer Institute; Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA.
2Department of Pediatrics, Division of Blood & Marrow Transplant & Cellular Therapy, and the Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA.
Address correspondence to: Bruce R. Blazar, University of Minnesota, Mayo Mail Code 366, 420 Delaware St SE, Minneapolis, Minnesota 55455, USA. Phone 612.626.1926; Email: blaza001@umn.edu.
Find articles by Kean, L. in: JCI | PubMed | Google Scholar
1Division of Pediatric Hematology and Oncology, Boston Children’s Hospital and the Dana-Farber Cancer Institute; Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA.
2Department of Pediatrics, Division of Blood & Marrow Transplant & Cellular Therapy, and the Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA.
Address correspondence to: Bruce R. Blazar, University of Minnesota, Mayo Mail Code 366, 420 Delaware St SE, Minneapolis, Minnesota 55455, USA. Phone 612.626.1926; Email: blaza001@umn.edu.
Find articles by Blazar, B. in: JCI | PubMed | Google Scholar
Published April 15, 2024 - More info
The Haitian proverb “Beyond mountains, there are mountains” has many meanings. One that resonates for hematopoietic stem cell transplantation (HCT) over the last 50-plus years is the sense that as you solve one problem, another presents itself. To mark the 100th anniversary of the JCI, we focus on the evolving understanding of major HCT hurdles, those surmounted, challenges that lie ahead (Figure 1), and how these have been chronicled through key JCI papers.
Mountain climbing: key advances in HCT and the unsolved challenges remaining. (A) Key accomplishments (left) and challenges remaining (right) for transplant preparation (3, 4), stem cell infusion (6, 7), and tissue repair after HCT (20, 21, 26). (B) Key accomplishments and challenges remaining for achieving immune homeostasis (11–18) and protecting against immune pathology after HCT (8–10, 22–29). References highlight major JCI articles contributing to the described discoveries.
Hematopoietic stem cell transplantation (HCT) is a curative therapy for many malignant and nonmalignant hematologic diseases, including treatment-refractory leukemia, classical hematologic diseases (e.g., sickle cell disease), congenital and acquired immune system disorders (e.g., severe combined immune deficiency), and inborn errors of metabolism (e.g., Hurler syndrome). Allogeneic HCT refers to a transplant where donor BM, peripheral blood–based stem cell, or umbilical cord blood products are infused into a patient after myeloablative or nonmyeloablative conditioning regimens involving irradiation and/or chemotherapy; these are designed to make room for donor stem cells, immune suppress the host to permit donor graft acceptance, and eliminate residual malignant cells. Autologous HCT refers to infusion of the patient’s own stem cells to restore hematopoiesis. Despite considerable successes, multiple challenges still loom. For allogeneic HCT, the focus of this Viewpoint, the greatest challenges are relapse of the primary disease and graft-versus-host disease (GVHD). Each deserves special discussion given their strong connection to the immunology of HCT, as well as to other HCT complications, including infection, graft failure, and organ failure.
For patients with malignant hematologic diseases, the transplanted donor hematopoietic stem and progenitor cells serve two purposes: (i) they replace the diseased BM; and (ii) they protect against relapse by what is known as the graft-versus-leukemia (GVL) effect, whereby transplanted immune cells, including both T cells and NK cells, provide immunologic surveillance and clearance of malignant cells. One of the greatest challenges of HCT is that the T cells that are so important for immune reconstitution and for GVL are also the major mediators of GVHD. Acute and chronic GVHD occur when T cells (along with other immune cells) fail to achieve immune tolerance to the tissues and organs of the recipient, become activated and pathogenic, and cause immune-mediated tissue destruction. During acute GVHD (aGVHD), immune cells cause inflammatory tissue damage to canonical organs (including the gastrointestinal [GI] tract, liver, and skin) along with other tissues (e.g., lung, CNS) (1). During chronic GVHD (cGVHD), T cells orchestrate a complex pathogenic program that also involves B cells and other immune mediators to cause sclerotic, fibrotic, and/or inflammatory damage, which can target any tissue in the patient and is the major cause of morbidity and mortality in patients surviving beyond 100 days after HCT (2).
A landmark 1959 article by Thomas et al. reported the first patients with leukemia given lethal doses of irradiation followed by HCT with bone marrow donated by the patients’ identical twins; this treatment overcame the alloengraftment barrier, circumvented GVHD barriers, and presciently highlighted critical antileukemia therapeutic needs (e.g., GVL effects) (3). Not until 1971 would serotyping and mixed lymphocyte culture testing of canine siblings confirm HLA matching and definitively show that without immune suppression, minor histocompatibility (miH) antigen mismatches pose a substantial risk of GVHD lethality (4), setting the stage for pursuing GVHD antigen discovery and predictive, diagnostic, and prognostic biomarkers (see below). Rapid, long-term hematopoietic recovery provided indirect evidence for chimerism and implied that bona fide donor hematopoietic stem cells (HSCs) repopulated the BM niche. Decades later, Ginsburg et al. deployed a sensitive, multilineage assay using DNA polymorphisms to quantify donor and host chimerism levels (5).
Many studies provided circumstantial evidence that the elusive HSC resided in a BM CD34+ subset. In a tour-de-force study using a cross-reactive nonhuman primate and human anti-CD34 mAb, Berenson et al. showed that CD34-enriched but not CD34-depleted cells are engraftable (6). The ability to manipulate CD34+ cells, a definition that became synonymous with HSCs, led to efforts to increase CD34+ cell numbers to achieve rapid hematopoietic recovery and to utilize this population for stem cell gene therapy. Cord blood CD34+ cells incubated with small-molecule nicotinamide, a primary precursor of nicotinamide adenine dinucleotide (NAD+) and cofactor in multiple redox reactions, showed promise for ex vivo HSC expansion, as reported in 2014 (7). With continued development, nicotinamide treatment secured FDA approval for stem cell therapy in 2023.
In a clinical trial, Bleakley, Shlomchik, and colleagues employed donor graft CD34+ selection supplemented with T cells depleted of GVHD-causing naive cells and enriched in memory cells that have the capacity to mediate antitumor and antimicrobial immunity (8). This strategy was hypothesized to decrease both aGVHD and cGVHD; cGVHD was virtually eliminated, but surprisingly, aGVHD frequency was unchanged. These data, indicative of the distinct biology of the two GVHD counterparts, offered a platform for cellular therapies under conditions wherein there is a prerequisite for enrolling cGVHD-free patients for safety and efficacy evaluation.
Despite improved HCT outcomes, GVHD remains a difficult mountain to climb. Notable successes and new hurdles have arisen with each advance. We cannot discuss GVHD prevention without first highlighting the seismic change in the field, based upon decades of pioneering studies by Johns Hopkins investigators, that incorporated posttransplant cyclophosphamide (PT-Cy) into a commonly used GVHD prophylaxis regimen. Adding PT-Cy to tacrolimus and mycophenolate mofetil (MMF) resulted in a striking decrease of cGVHD (9), similar to naive T cell depletion. Compared with the historic standard of care of a calcineurin inhibitor plus methotrexate for GVHD prophylaxis, PT-Cy/tacrolimus/MMF led to a significantly improved composite cGVHD survival endpoint (Bone Marrow Transplant Clinical Trials Network trial [BMT CTN 1703]).
It has been challenging to rigorously pin down the mechanism(s) by which PT-Cy prevents cGVHD in patients. Furthermore, there are substantial trade-offs with this therapy, including the cardiac toxicity inherent to cyclophosphamide, as well as adverse effects on T cell reconstitution. Toward understanding both the benefits and risks of PT-Cy, Kanakry’s group has pursued elucidation of the mechanisms by which PT-Cy prevents GVHD in mice (10). They found that GVHD prevention by PT-Cy involved complex immunologic mechanisms including T cell dysfunction and increased suppression associated with rapid reconstitution of Tregs. Whether these mechanisms can be observed in humans is an open question and one that is currently being answered through the BMT CTN 1801 biology study (ClinicalTrials.gov NCT03959241).
Rather than nonspecific removal or incapacitation of donor T cells, a long-standing GVHD prophylaxis goal has been the targeted inactivation of GVHD-causing T cells. Blockade of T cell costimulatory signals, designed to selectively prevent alloreactivity while retaining protective immunity, inspired our group to investigate multiple costimulatory pathways, with a special focus on CTLA-4–Ig (abatacept) and anti-CD28 mAb blockade of CD28-CD80/86 costimulation (11). These studies provided an important counterpart to the PT-Cy story, wherein abatacept has proven highly effective in preventing aGVHD, but not cGVHD (at least with a 4-dose regimen). Abatacept’s substantial effects of decreasing aGVHD and improving survival after unrelated-donor HCT led to its being the first FDA-approved GVHD prevention strategy. A 4- versus 8-dose randomized trial geared toward preventing both aGVHD and cGVHD (ClinicalTrials.gov NCT04380740) underscores the critical need for clinical trials to investigate tradeoffs we now face to identify regimen(s) that functionally controls aGVHD and cGVHD without physical T cell depletion.
A promising partner to CD28-80/86 blockade is the CD40:CD154 pathway. A classic paper by Blazar et. al first documented anti-CD154 mAb–induced pre-HCT donor CD4+ T cell alloantigen-specific tolerization and protective immunity retention (12). Long-lived, effective anti-CD154 mAbs that do not cause unwanted thrombotic complications now exist and have advanced to clinical trials for ameliorating autoimmune disease and preventing solid organ graft rejection.
As the field advances with increasingly efficacious therapeutics, the quest to understand mechanisms causing GVHD and GVHD-inciting host antigens continues. Korngold found that CD4+ T cell allorecognition of multiple miH antigens exclusively presented on hematopoietic cells resulted in self-limited aGVHD, whereas lethal aGVHD was induced by nonlymphoid miH antigenic disparities (13). Maillard’s lab added to this puzzle by proving that GVHD-amplifying signals can be delivered by host fibroblastic stromal cells including fibroblastic reticular cells (FRCs) that express Notch ligands and are juxtaposed to alloreactive T cells (14). As described by Chakraverty et al., GVHD downregulated expression of the autoimmune regulator–like transcription factor DEAF1 — necessary for intranodal peripheral tissue–restricted self-antigen display and prevention of autoimmunity — in FRCs. Ultimately, FRC depletion by GVHD may account for the acute-to-chronic GVHD transition (15).
A major step forward in our understanding of cGVHD mechanisms was provided by Ritz and colleagues, who observed inadequate peripheral reconstitution of naive Tregs caused by GVHD-induced thymus injury, resulting in a predominance of proliferating activated/memory Tregs with high susceptibility to Fas-mediated apoptosis and failed cGVHD control of autoimmune-like manifestations (16). Simultaneous with studies by Edinger et al. (17), Cohen’s lab showed that recipient-specific Tregs not only controlled GVHD and GVL effects but facilitated immune reconstitution (18).
As the field of GVHD mechanistic studies has matured and new strategies are sought to continue to perfect GVHD prophylactic regimens, the focus of the field has turned increasingly to understanding drivers of tissue injury. Contrary to the long-accepted paradigm that donor T cells alone cause aGVHD tissue destruction, Kupper and colleagues published their finding that proliferating, proinflammatory T cells that were 100% of host origin could be found adjacent to donor antigen-presenting cells (APCs) in patient with skin aGVHD, consistent with host skin-resident T cell activation by donor APCs and subsequent tissue injury (19). Key data pertinent to GI tract injury are findings of increased levels of circulating regenerating islet-derived protein III-gamma (Reg3γ), an antimicrobial peptide released by damaged intestinal Paneth cells, coincident with this deadly GVHD manifestation (20). Such observations led to the administration of IL-22, produced by gut innate lymphoid cells type 3 (ILC3s) to stimulate nonhematopoietic epithelial and stromal cell proliferation, host defense at barrier surfaces, and intestinal stem cell proliferation, resulting in increased Paneth cell and decreased Reg3γ levels.
Antiinflammatory gut ILC2s can ameliorate GI tract damage, as shown by Bruce, Serody, and colleagues, who demonstrated marked intestinal ILC2 depletion following a pre-HCT conditioning regimen that required at least 3 months for repopulation (21). Conversely, the infusion of donor ILC2s, isolated from the peritoneum, mitigated aGVHD lethality due at least in part to IL-13–stimulated myeloid-derived suppressor cells.
As allo-HCT has become safer overall, clinical trials have increasingly included older patients who receive peripheral blood stem cell grafts. However, these transplant paradigms can increase cGVHD risk, which emphasizes the need for continued preclinical investigations into the mechanisms of cGVHD prevention and treatment as a prelude to clinical translation. Collaborative studies between the Blazar and Byrd laboratories showed that the dual Bruton’s tyrosine kinase (BTK)/IL-2–induced T cell kinase (ITK) inhibitor ibrutinib could reduce murine cGVHD, including in preclinical models of often treatment-refractory lung disease (bronchiolitis obliterans) and sclerotic skin manifestations (22). A phase I ibrutinib trial in patients with steroid-refractory cGVHD designed to preclude pathogenic B cell–T cell cooperativity led to rapid FDA approval for this debilitating disease. Other FDA-approved immunomodulatory drugs for steroid-refractory or -resistant cGVHD include inhibitors of the Rho-associated coiled-coil containing protein kinase 2 (Rock-2; belumosudil) (23) and JAK1/2 (ruxolitinib) (24).
Groundbreaking studies by MacDonald, Blazar, Hill, and colleagues identified another major cellular contributor to cGVHD: profibrotic macrophages recruited to the skin and the lung, regions with antibody deposition and tissue injury, and implicated in scleroderma-like skin and lung bronchiolitis obliterans (25). Neutralizing anti-CSF1R mAb efficacy in cGVHD mice has now been successfully translated into the clinic to treat refractory cGVHD (ClinicalTrials.gov NCT04710576).
One of the major hurdles that remains in our field is the development of less-toxic but highly effective targeted approaches to engraftment during HCT. Toward this goal, the DiPersio lab has described antibody-drug conjugates (ADCs) against CD45 and CD117 that are able to effectively clear the BM and enable alloengraftment across MHC barriers, a major advance (26). When ADCs were combined with JAK1/2 inhibitors and BM ablation, alloengraftment across MHC barriers was achieved, GVHD was ameliorated, GVL was maintained, and thymic function was preserved, highlighting the next horizon of combined targeted conditioning and immune modulation for optimal patient safety and efficacy.
Strategies to control aGVHD and cGVHD, restore immune homeostasis, and minimize myeloablative regimen toxicity, each can contribute to an increased risk of relapse after HCT. On the other hand, proactive approaches to remove GVHD-causing donor graft T cells while sparing donor cells that possess GVL properties (8) or administering GVHD prophylaxis or treatment agents that spare steroids can inhibit aGVHD (26) and cGVHD (22) and even eradicate leukemia cells, thereby actually augmenting GVL. Toward these goals, seminal studies by the Murphy laboratory demonstrated that after infusion, activated NK cells had the anti-GVHD and pro-GVL effects of killing tumor cells and, via TGF-β release, mitigating aGVHD (27). Subsequently, Romee and colleagues infused in vitro generated cytokine-induced memory-like NK cells and reported a rapid, 10- to 50-fold expansion and increased in vivo cytolytic potency, along with a reduction in the level of malignant myeloid-lineage cells in 4 of 6 patients (28). Betts and Davila eradicated human CD83+ pathogenic CD4+ T cells, proinflammatory DCs, and AML cells by redirecting donor T cells to express an anti-CD83–expressing chimeric antigen receptor (29), adding another dual-purpose immune-based GVHD and GVL strategy. These examples highlight the risks, and new potential solutions, to the major problem of malignant disease relapse that continues to loom after transplant.
Though we have yet mountains to climb, this 50-year journey, viewed through the lens of JCI publications, illustrates remarkable progress and serves as a guidepost and North Star for future accomplishments.
We are grateful to the many patients, family members, colleagues, and trainees who have been instrumental in facilitating the remarkable evolution of HCT. Due to limitations in allowable numbers of references and words, we were unable to include numerous highly meritorious articles that contributed invaluably to the extraordinary progress observed in HCT over the past 50 years. This work was supported in part by NIH/National Institute of Allergy and Infectious Diseases (NIAID) grants U19 Al1051731, U19 AI174967; NIH/National Heart, Lung, and Blood Institute (NHLBI) R01 HL095791, P01 HL158504, P01 HL158505; NIH/FDA R01 FD004099; a Leukemia and Lymphoma Society Translational Research Program Grant; and A CURE Childhood Cancer Grant (to LSK); and NIH/NIAID R37 AI34495, P01 AI056299; NIH/NHLBI R01 HL11879, R01 HL155114, R01 HL 147324, P01 HL158805, and NIH/National Cancer Institute (NCI) P01 CA065493 grants (to BRB).
Address correspondence to: Bruce R. Blazar, University of Minnesota, Mayo Mail Code 366, 420 Delaware St SE, Minneapolis, Minnesota 55455, USA. Phone 612.626.1926; Email: blaza001@umn.edu.
Conflict of interest: LSK is on the scientific advisory board for Mammoth Biosciences and HiFiBio. She reports research funding from Magenta Therapeutics, Tessera Therapeutics, Novartis, EMD Serono, Gilead Pharmaceuticals, and Regeneron Pharmaceuticals. She reports consulting fees from Vertex; and grants and personal fees from Bristol Myers Squibb. LSK’s conflict of interest with Bristol Myers Squibb is managed under an agreement with Harvard Medical School. BRB reports research funding from BlueRock Therapeutics and Carisma Therapeutics and consulting fees from BlueRock Therapeutics, Editas Medicine, Janssen Oncology, Sandoz, Legend Biotech, GentiBio Inc., and Magenta Therapeutics.
Copyright: © 2024, Kean et al. This is an open access article published under the terms of the Creative Commons Attribution 4.0 International License.
Reference information:J Clin Invest. 2024;134(8):e179944. https://doi.org/10.1172/JCI179944.
Solid organ transplantation: solid but not yet spectacularLaurence A. Turka
Leptin physiology and pathophysiology: knowns and unknowns 30 years after its discoveryJeffrey S. Flier et al.
The GLP-1 journey: from discovery science to therapeutic impactDaniel J. Drucker
CAR T cells for hematological malignanciesBarbara Savoldo et al.
Clinical investigation of hypoxia-inducible factors: getting thereGregg L. Semenza
Pancreatic β cell function versus insulin resistance: application of the hyperbolic law of glucose toleranceRichard N. Bergman
Nitric oxide in vascular biology: elegance in complexityJoseph Loscalzo
How the JCI’s most-cited paper sparked the field of lipoprotein researchMichael S. Brown et al.
SGLT2 inhibitors: cardiorenal metabolic drugs for the agesRalph A. DeFronzo
G protein–coupled receptors: from radioligand binding to cellular signalingHoward A. Rockman et al.
Bisphosphonates for osteoporosis: from bench to clinicTeresita Bellido
Pandemics past, present, and future: progress and persistent risksArturo Casadevall
Life-saving effect of pulmonary surfactant in premature babiesJ. Usha Raj et al.
Advancing chemokine research: the molecular function of CXCL8Yiran Hou et al.
Navigating an enigma: the continuing journey of autoimmunity discoveriesMariana J. Kaplan
Defining and targeting mechanisms of eosinophilic inflammation in a new era of severe asthma treatmentJoshua A. Boyce et al.
The other pandemic: lessons from 40 years of HIV researchMary E. Klotman et al.
A half-century of VEGFA: from theory to practiceSusan E. Quaggin
Two decades of advances in preeclampsia research: molecular mechanisms and translational studiesS. Ananth Karumanchi
The forgotten pandemic: how understanding cholera illuminated mechanisms of chloride channels in multiple diseasesQais Al-Awqati
Is it time to rethink the relationship between adipose inflammation and insulin resistance?Evan D. Rosen et al.
Checkpoint therapy in cancer treatment: progress, challenges, and future directionsMesude Bicak et al.
The arc of discovery, from the description of cystic fibrosis to effective treatmentsMichael J. Welsh
Intertwining clonality and resistance: Staphylococcus aureus in the antibiotic eraHenry F. Chambers et al.
The expanding application of antisense oligonucleotides to neurodegenerative diseasesCharlotte J. Sumner et al.
Toward a better understanding of chronic hepatitis B virus infectionBarbara Rehermann
The convergence of genomic medicine and translational omics in transforming breast cancer patient careSulin Wu et al.