Cellular therapies play a major and expanding role in the treatment of hematologic diseases. For each of these therapies, a narrow therapeutic window exists, where efficacy is maximized and toxicities minimized. This review focuses on one of the most established cellular therapies, hematopoietic stem cell transplant, and one of the newest cellular therapies, chimeric antigen receptor–T cells. In this review, I will discuss the current state of the field for clinical end point analysis with each of these therapeutics, including their critical toxicities, and focus on the major elements of success for each of these complex treatments for hematologic disease.

The scope of cellular therapeutics is broad and expanding. Although adoptive T-cell therapeutics (in particular, chimeric antigen receptor T [CAR-T] cells) have garnered the lion’s share of attention in the past several years, these newly US Food and Drug Administration (FDA)-approved therapeutics still represent a small proportion of the entire scope of cellular therapies in clinical use, and those undergoing clinical investigation for patients with hematologic diseases. Indeed, the cellular therapy in widest use remains hematopoietic stem cell transplant (HCT), with many new initiatives in graft manipulation designed to increase efficacy and decrease transplant-associated complications. In addition to CAR-T therapies, there are also a number of other T-cell therapies that do not involve CARs (most prominently including antiviral T cells as well as T-cell therapeutics using tumor-specific T-cell receptors), as well other effector cells (particularly natural killer cells) designed to reduce malignant relapse. Finally, there are the regulatory cellular therapies, most prominently including CD4+FoxP3+ regulatory T cells, as well as a number of other suppressive cell-based therapies (including mesenchymal stromal cells, myeloid-derived suppressor cells, and type 1 regulatory T cells, among others). For each of these therapies, a critical therapeutic window exists, where efficacy is maximized and toxicities minimized. This review will focus on HCT and CAR-T-cell therapies, in which this therapeutic window can be small, and in which toxicities, when they occur, can be fatal. It will provide an overview of the state of the art in monitoring clinical efficacy and toxicity for each, and the surrogate markers that are most useful for interrogating their attendant risk-to-benefit ratios.

HCT is a complex procedure with multiple stages, and at each stage, critical elements exist that define success or failure. It is useful to divide the transplant process into 4 phases: (1) transplant conditioning; (2) day of transplant; (3) preengraftment; (4) postengraftment. The gold standard of transplant success, encompassing success at all of the stages, is a patient who, a year posttransplant (and thereafter), is alive and in remission and has successfully reconstituted protective immunity without graft-versus-host disease (GVHD). The key clinical end points that encompass each of these milestones and the accompanying surrogate markers (if they exist) are discussed in the following sections.

Transplant conditioning

Conditioning for transplant can be divided into regimens that are expected to completely (or nearly completely) ablate the recipient marrow (“myeloablative”) and those that would be considered nonmyeloablative and thus are not expected, in themselves, to ablate the marrow. Of the myeloablative-conditioning strategies, some are less toxic than others, with these less-toxic strategies deemed “reduced-intensity” or “reduced-toxicity” conditioning, in contrast to the standard myeloablative total body irradiation– and high-dose chemotherapy–based strategies. Although conditioning is a critical component to transplant, there are no standard surrogate markers for conditioning success, other than the eventual engraftment of donor marrow. However, a robust comorbidity index exists that helps to stratify patients for their risk of transplant-mediated toxicities and for transplant-related mortality, thereby informing decision-making about conditioning intensity.1,2  The well-described impact of comorbidities on nonrelapse mortality (NRM) and the increasing average age of patients undergoing HCT (as well as the well-recognized increase in NRM in patients younger than 1 year of age who are often transplanted for genetic diseases) are driving research into more targeted, less globally toxic conditioning regimens. One of the most exciting strategies involves the use of targeted monoclonal antibodies, most prominently those targeting c-kit (CD117), CD47, and CD45, studies of which have recently been published by groups at the Fred Hutchinson Cancer Research Center,3-5  Stanford University,6  and Harvard University.7  Although these antibodies are not specific for hematopoietic stem cells, off-target toxicity has not been a major problem to date. Although clinical trials using these new agents are just beginning, the advent of these antibody-based approaches represents a major advance, and ushers in a new era of nongenotoxic transplantation.

Day of transplant

Transplant day (day 0) is the shortest phase of every transplant, but potentially the most important in terms of determining transplant success or failure. The prevailing graft sources (bone marrow, peripheral blood stem cells, cord blood) and the spectrum of HLA matching all make a major impact on transplant outcome, but are beyond the scope of this review. This review will focus on graft engineering, as it is this maneuver that ties HCT most closely with other cellular therapies.

One of the major strategies for graft manipulation, which has been used for several decades, is T-cell depletion. Ex vivo T-cell depletion was first applied to many graft types in the 1980s,8-18  and gained traction with HLA-haploidentical HCT, given the significant risk of severe GVHD that occurred with haplo-HCT in the pre-posttransplant cyclophosphamide era.19-22  This strategy was initially performed by removal of T cells from the graft,23  and is now accomplished by positive selection of CD34+ cells.24  Although this strategy has been highly effective in controlling GVHD, other complications, including graft loss, malignant relapse, and infectious complications remain significant issues, although there are several important studies from groups in Perugia, Italy, and at the Memorial Sloan Kettering Cancer Center (MSKCC) in which relapse rates were not increased despite the control of both acute and chronic GVHD.22,25-27 

Given the ongoing concerns with whole-scale T-cell depletion (especially without high-intensity pretransplant conditioning), recent work has concentrated on a more nuanced approach, which involves removal of the αβ+ T cells28,29  (those that are most implicated in alloreactivity) while preserving γδ T cells, natural killer cells, monocytes, and dendritic cells,28-30  thus better preserving antimicrobial protective immunity and graft-versus-leukemia.31-34  Another approach that has been added to the αβ−T-cell depletion strategy has been the gene modification of T cells by the gene encoding caspase-9, iC9,35,36  which renders them susceptible to ablation with the dimerizing agent AP1903,36-39  and the subsequent addition of titrated amounts of these ablatable T cells posttransplant.40  Initial results have been encouraging41  and a number of larger trials are ongoing to fully evaluate this approach. The final strategy for graft engineering that will be discussed is naive T-cell depletion. This approach was based on substantial preclinical data supporting the critical role that naive T cells play in GVHD in murine studies.42-44  Based on these results, several groups have begun clinical investigations using this strategy.45-48  The results from the Fred Hutchinson Cancer Research Center have documented a striking decrease in chronic GVHD despite no substantial impact on acute GVHD when naive T-cell–depleted peripheral blood stem cells were transplanted after a high-intensity conditioning regimen.47,48  The impact on chronic GVHD was encouraging, and larger, multicenter trials of this graft-engineering approach are currently under way.

Preengraftment, hematologic engraftment, and immune reconstitution

In the preengraftment period, there are a number of significant risks, associated both with the ongoing toxicities of conditioning and with the inherent risks of pancytopenia. In this phase of transplant, the surrogate markers are well established, and include the day of neutrophil engraftment, the day of platelet engraftment, and the degree of donor chimerism (with day 30 assessments standardly performed). Although the concept of successful engraftment has historically been associated with the rise in neutrophil count (and its attendant decrease in the risk of serious bacterial and fungal infection), there has been less certainty about what constitutes fully functional hematologic and immunologic reconstitution. In particular, surrogate markers for successful immunologic reconstitution, as it relates to lymphocyte engraftment, antiviral immunity, and the impact on late-onset immune dysregulation have not yet been established. Although historically a CD4 count of >200 × 103 cells per μL had been implicated as at least partially protective against some viral infections,49  our understanding of the pace and character of effective immune reconstitution is becoming more comprehensive, as has been our understanding of the impact that latent viruses can make on this reconstitution. Thus, recent work from my laboratory and others in both unrelated donor and haploidentical HCT has documented the significant quantitative and qualitative effect that cytomegalovirus (CMV) reactivation can make on posttransplant T-cell expansion.50,51  Of note, this work has underscored the fact that, although CMV is the major driver of the quantitative reconstitution of effector CD8 cells, this does not necessarily equate to functional competence. Indeed, our calculations of the number of deficits in the T-cell repertoire in patients who reactivated CMV compared with those who did not suggest that CMV-reactivating patients may have significant defects in T-cell–mediated protective immunity.50  This observation is consistent with a recent study from MSKCC, which focused on T-cell–depleted HCT, and demonstrated that functional competence of T cells after mitogen stimulation is more predictive of survival advantage after HCT than quantitative lymphocyte expansion.52  This functional competence, and the diversity of the T-cell repertoire, can also be linked to thymic health after transplant.53-59  The surrogate markers for this competence are also becoming more sophisticated and include T-cell receptor excision circle assays, naive T-cell regeneration, and T-cell receptor repertoire diversity in both naive and antigen-experienced CD4 and CD8 T-cell populations.54-59 

Postneutrophil engraftment

The period after neutrophil engraftment and through the first year posttransplant is dominated by 2 major toxicities, each of which there has been the subject of major initiatives to identify surrogate markers to help guide preemptive and treatment strategies. The first toxicity, GVHD, represents the major cause of NRM for the vast majority of transplant paradigms. Although clinical staging and grading of both acute and chronic GVHD are well established, our ability to predict and develop accurate prognoses for patients with GVHD is still in its infancy. Given the importance of preventing and treating GVHD, over the past decade, there has been a major emphasis on developing biomarkers for this disease. For acute GVHD, much progress has been made in identifying a panel of serum biomarkers that can risk-stratify patients who develop acute GVHD.60-73  This work has now matured to the point where clinical trials can incorporate biomarkers as well as sophisticated clinical staging74  to risk-stratify patients for treatment studies, representing a major step forward for the field. The field of chronic GVHD is also making strides in the areas of soluble biomarkers,75-85  although these are not yet as robust as those for acute GVHD, likely due to the inherent complexity of chronic GVHD, in terms of its timing, clinical presentation, and underlying immunopathologic drivers. There is also a growing focus in the field in identifying cellular biomarkers of this disease. This work has been driven by detailed flow cytometric evaluation of the T- and B-cell subpopulations driving both acute and chronic disease,86-102  as well as increasing use of systems-based transcriptomic approaches to identifying the pathways associated with GVHD, both in preclinical models and clinical samples.50,103-107  This work is identifying a new cohort of targetable pathways for GVHD control, many of which are amenable for clinical translation.

Although GVHD is the most significant cause of NRM after HCT, the primary cause of death after transplant for patients with leukemia or lymphoma remains relapse of their primary disease. The issue of disease relapse, and the development of strategies to prevent and/or treat relapse has undergone an explosion of activity in the 5 five years, based on the landmark success of cellular therapies designed to eliminate malignancies. The most striking successes have been with the CAR-T-cell therapies targeting the CD19 antigen. However, as with other cellular therapies, CAR-T cells are associated with a complex risk-to-benefit profile.

For patients with relapsed leukemia, and for those with primary refractory disease, the outlook with conventional chemotherapy remains dismal. In the past 5 years, a subset of these patients, predominantly including those with CD19-expressing acute lymphoblastic leukemia (ALL), have been successfully treated with CD19-redirected CAR-T cells, which has represented a watershed moment for cellular therapeutics. This success has been built on decades of research and the development of both immune-engineering and gene transfer capabilities, with the first successful trials published in pediatric B-cell ALL in 2013,108  with multiple follow-up studies now completed. The striking efficacy of this therapeutic strategy has led to the rapid approval of 2 CAR-T therapies: tisagenlecleucel and axicabtagene ciloleucel, with more approvals expected. Although the vast majority of CAR-T-cell trials for hematologic diseases have focused on CD19-redirected T cells, there are a number of new trials targeting other B-cell antigens (including CD20, CD22, CD30, and B-cell maturation antigen109-112 ), T-cell antigens, as well as early trials targeting antigens associated with acute myeloid leukemia.113-124 

As with HCT, CAR-T-cell therapies are composed of multiple stages, and at each stage, critical elements exist that contribute to success or failure. It is useful to divide CAR-T cellular therapies into 4 distinct stages: (1) patient selection and cell manufacturing; (2) preinfusion chemotherapy; (3) remission-induction and CAR-T–associated toxicities; and (4) postremission therapeutic strategies. Given the early stage of the field of CAR-T cellular therapeutics, there is still considerable debate about what constitutes success in this field. Thus far, the definition of success has focused on remission induction, with many patients receiving CAR-T cells going on to have further consolidative therapy (most commonly including HCT). Indeed, the reimbursement strategy for the first FDA-approved CD19-CAR-T product, tisagenlecleucel, includes charging for this therapy only if successful remission (but not long-term cure) is achieved. However, as CAR-Ts are more widely used, and used earlier in the treatment pipeline, these definitions of success will need to evolve, and critical questions about the acceptability of CAR-T–associated toxicities, and the need for additional treatment after CAR-T therapy (and how to select patients that do or do not need further treatment) will need further refinement.

Cell manufacturing and patient selection

Although there is growing interest in the development of universal, “off-the-shelf” CAR-T products,125,126  the prevailing paradigm for CAR-T-cell manufacture currently relies on the creation of a patient-specific product. To manufacture this product, T cells need to be obtained from the patient (usually by apheresis) and then expanded ex vivo to reach the desired infusion dose. This process can be challenging, especially in the heavily pretreated patient populations currently being treated with CAR-Ts. Although the first studies of CAR-Ts included a relatively high rate of failure to successfully manufacture a CAR-T product (and included a preexpansion feasibility assessment, which eliminated significant numbers of prospective patients),127  newer studies have documented high rates of success (>95% for ALL and >89% for neuroblastoma in the largest study of manufacturing efficacy to date).128 

The composition of CAR-T-cell products has also evolved, although there is not yet a recognized gold standard. Two key elements of CAR-T composition need to be considered: (1) the structure of the CAR construct itself, and (2) the cellular composition of the infused product. (1) The structure of the CAR transgene has evolved from a “first-generation” structure, which expressed the CD3-ζ signaling domain, to “second-generation” CARs, in which one of many possible costimulatory signals (the 2 most common being CD28 and 41BB signals) were added to CD3-ζ, to “third-generation” CARs, which contain 2 costimulatory signals in tandem (CAR construct structure reviewed in June et al129 ). Although the identity of the costimulatory domains have gained the most attention of late, there are multiple other considerations in engineering an optimal CAR construct, and a comprehensive discussion of these is beyond the scope of this review. These engineering considerations have been recently reviewed by Srivastava and Riddell, Gomes-Silva and Ramos, and Lim and June.130-132  (2) The cellular composition of the CAR-T-cell infusion is similarly complex and is also rapidly evolving. The original CAR-T infusions contained unselected products from expansion cultures; thus, patient-specific variability in terms of CD4:CD8 ratio, naive/central/effector memory composition, and the proportion of CAR-T–transduced cells ensued. Given the enhanced ability of central memory T cells to expand and persist after adoptive transfer,133-136  several studies have enriched CAR-T products for this subpopulation. Although current CAR-T infusion strategies often do not include the purification of specific memory subpopulations, CAR-T culture conditions optimized separately for CD4 and CD8 expansion have been developed that are also geared toward producing a cell product with a defined CD4:CD8 ratio, and with optimized expansion and persistence characteristics.137,138  Although many strategies for defining and optimizing CAR-T products exist, a gold standard for this aspect of CAR-T preparation has not been established, with wide variation in the culture conditions and final composition of CAR-T-cell products. It is also important to consider the fact that in any CAR-T production process, not all T cells are successfully transduced with the CAR-T construct. Therefore, most manufacturing processes now include enrichment and/or selection for the transduced cells, such that the infused product does not contain a surfeit of non-CAR-Ts. Importantly, although boutique manufacturing processes have, in the past, been able to handle the demand for CAR-Ts, as the number of products needed grows, more industrial, “untouched” manufacture processes will be required, and creating a system for CAR-T manufacturing that can more closely resemble a blood bank than a research laboratory (without compromising efficacy) will be a critical area of future development.

Preinfusion chemotherapy

To optimize CAR-T-cell expansion after infusion, a state of lymphodepletion is usually induced prior to cell delivery. This approach capitalizes on the well-documented phenomenon of homeostatic T-cell proliferation, which leads to T-cell expansion, activation, and memory differentiation.139-141  Initial studies often used cyclophosphamide alone for lymphodepletion, but enhanced expansion and persistence has been documented when cyclophosphamide is combined with other agents, most commonly including fludarabine. Although there has been some concern that fludarabine may have increased the risk for neurotoxicity (given its known association with neurologic events at very high doses), which is observed after CAR-T therapy (discussed in “CRES”), the predominance of the data argues against this association being causative,142,143  and cyclophosphamide/fludarabine chemotherapeutic regimens are widely used with both CD19-directed and other CAR-T therapies. The goal of preinfusion chemotherapy is twofold: (1) to deplete endogenous T cells that might increase the risk of T-mediated rejection of CAR-T cells,144  and (2) to enhance CAR-T expansion in the lymphodepleted host.139-141 

Remission induction and associated toxicities

One of the most remarkable results observed with CAR-T-cell therapies against B-cell malignancies has been the high rates of complete remission (CR) that have been observed (over 90% in some studies),108,136-138,144-155  which are particularly striking given the high-risk patient populations that have been treated. However, despite the successful remission induction with CD19 CAR-Ts, these cells have also been associated with significant toxicities. These include both a cytokine release syndrome (CRS) and a neurotoxicity syndrome (newly termed CAR-T–related encephalopathy syndrome [CRES]). CRS was the first major toxicity measured in patients treated with CD19-CAR-T cells, and remains the most commonly observed toxicity.108  This entity encompasses a large number of signs and symptoms, which range from low-grade fever and constitutional symptoms to life-threatening multiorgan dysfunction, high fever, and hypotension. Neurotoxicity, or CRES, is the more rare, but most deadly complication of CAR-T cells, and is characterized by confusion, delirium, language disturbance, seizures, and cerebral edema. Given the potential severity of both of these complications, there have been significant efforts made in recent years in defining these toxicities and testing potential prevention or treatment modalities, including the formation of toxicity working groups156  to define cross-institution standards for diagnostic criteria and treatment algorithms. Although significant advances have been made, universal consensus has not been reached,157  and remains a critical unmet need in the field, especially in the setting of FDA approval, as these therapies move from specialized centers to more broad implementation.

CRS.

CRS begins with the activation of T cells, when the CAR engages its cognate antigen on both malignant and nonmalignant cells. The active mechanisms causing CRS include the release of cytokines and chemokines by the CAR-Ts themselves (prominently including interleukin-6 [IL-6], IL-2, soluble CD25, interferon γ) along with activated “bystander” immune cells (including monocytes and macrophages) that secrete multiple inflammatory mediators.137,152,158-161  Although all of the risk factors for severe CRS have not been determined, patients at higher risk have been found to have higher CD19 antigen load (either from disease or normal B cells), and to develop CRS earlier after cell infusion (usually <3 days after infusion138,152,161,162 ). Surrogate markers for severe CRS that are both sensitive and specific are still being elucidated, with these studies garnering increasing statistical power and predicative capability as increasing numbers of patients are treated with CAR-T cells. Two robust correlative markers of severe CRS have been (1) the degree of expansion (and the peak levels) of CAR-T cells measured in the peripheral blood and (2) the presence of highly elevated serum IL-6 levels.137,138,142,152,156,159  Moreover, the functional association of IL-6 levels with clinical disease has been demonstrated by the ability of the anti-IL6R antibody tocilizumab (FDA approved for CAR-T-cell therapy in 2017) and the anti-IL-6 monoclonal antibody siltuximab to effectively treat symptoms of CRS.138,152,162-165  In addition to IL-6 blockade, corticosteroids have also been used to treat CRS,163,165,166  but their use has been viewed with caution, given concerns that this treatment might blunt the antileukemia efficacy of CAR-T-cell therapy. Despite the understandable concerns, several studies have observed that use of steroids did not appear to affect CR rates nor durability of CAR-T cells, although extensive follow-up has not yet been completed.138,156,159,160 

CRES.

Less prevalent than CRS, but clinically more concerning, is CRES, the toxic encephalopathic syndrome that can accompany CD19 CAR-T therapy.137,142,144,155,167,168  Although often self-limiting, the syndrome can also be severe, and result in seizures, obtundation, increased intracranial pressure, and cerebral edema, which has, in a small proportion of patients, led to death.137,142,144,155,161,167-169  Given the severity and potential morbidity/mortality associated with CRES, there has been a significant effort made to understand its pathophysiology. Several hypotheses have been put forth. The first focuses on the inflammatory cytokine milieu that accompanies CD19 CAR-T therapy, and the accumulation of these cytokines in the brain in the setting of high serum levels.151,156,159,160,165  The second focuses on direct T-cell infiltration into the cerebral spinal fluid (CSF) and brain.108,151,152,165,170  Compelling new data also implicate breakdown of the blood-brain barrier (BBB) in CRES, supported by both histopathologic evidence and by the measurement of systemic biomarkers for endothelial disruption during CRES.142,168  A recent study by Gust et al has proposed a pathophysiologic model of the interplay between CAR-Ts, cytokines, the BBB, and CRES.142  In this model, the inflammation associated with CRS leads to activation of endothelial cells in the central nervous system (CNS), which drives release of 2 key mediators of the ensuing BBB disruption. These are ANG2 and von Willebrand factor, which together drive endothelial cell activation and BBB disruption as well as the coagulopathy that is often observed during CRES. The BBB disruption results in a “feed forward” loop in which more cells and cytokines can cross into the CNS, leading to further activation of endothelial cells and thus further CNS inflammation. This has important clinical implications, as it suggests that therapeutic strategies that restore normal ANG2 or von Willebrand factor levels may be able to prevent or treat CRES. Given the concern that corticosteroids may impair long-term CAR efficacy, finding more targeted agents, such as those that target ANG-2, would be a major advance for the field.

Although the clinical studies are now starting to yield important clues to the pathophysiology of CRES, one of the major barriers to understanding the molecular pathobiology of CAR-T–mediated neurotoxicity has been the lack of animal models for this disorder. To address this, my research group, in collaboration with Michael Jensen’s laboratory, has recently developed the first nonhuman primate (NHP) model of CRS and neurologic toxicity, using CD20 CAR-T cells in rhesus macaques171  and Bruce Blazar’s group has recently developed a mouse model of CAR-T toxicity, in which human CD19-specific mouse CAR-T cells were adoptively transferred into mice whose normal B cells express a hCD19 transgene at hemizygous levels.172  Using the NHP model, we demonstrated CAR-T-cell expansion and B-cell aplasia, as well as CRS and neurotoxicity that closely mirrors what has been observed clinically.171  Thus, this model induces elevations in the serum of multiple cytokines, and has documented disproportionately high concentrations of several cytokines in the CSF. Importantly, it has also been able to recapitulate clinical and histopathologic neurotoxicity. Coincident with the clinical neurotoxicity, we identified significant encephalitis, which was characterized not only by the accumulation of CAR-T cells, as expected, but also by the accumulation of non–CAR-T cells that infiltrated both the CSF and brain parenchyma. The results of the NHP model and of the new clinical studies suggest that neurotoxicity is associated with a complex program of immune activation, which encompasses multiple cellular and soluble mediators, which include (1) an increase in multiple cytokines in the CSF compared with the serum and (2) the development of encephalitis, in which both CAR and non–CAR-T cells accumulate in both the CSF and the brain. The NHP model thus also suggests that the breakdown in the integrity of the BBB is key to clinical neurotoxicity and that strategies designed to protect this barrier may be key to protecting patients from this major complication of CAR-T-cell therapy.

Postremission therapeutic strategies

One of the key unanswered questions in CAR-T-cell therapeutics, even for the most efficacious CD19-CAR-T products, is how best to manage patients after successful remission induction. The first wave of studies enrolled patients who were at very high risk of imminent death from their primary disease: these patients had been refractory to standard treatment approaches or had relapsed (often multiple times). They had very few, if any, alternatives among the more conventional therapeutic approaches. In these patients, the achievement of a CR was a major achievement. However, although the successful induction of CR is critical, this is not, in itself, a sufficient end point to determine the ultimate success or failure of CAR-Ts. Longer follow-up, focused on the stability of CR for years after infusion, is beginning to be reported and provides both reason for celebration and also a mandate for further optimization of these therapeutics.138,144,152,173  Thus, recent work from multiple centers in the United States and China have documented high CR rates as well as sustained remissions in patients treated with CD19-CAR-T cells, both with and without additional consolidation,108,111,136-138,144-155,173  striking results given the high-risk patient populations that have been treated. However, it is now clear that for most patients, the CR is not followed by long-term remission, with more than half of patients ultimately relapsing following CAR-T therapy. Thus, several recent studies continue to document high (∼70% to 90%) remission rates in pediatric and adult patients with B-cell ALL,144,145,151,152,162,174-176  but with significant relapse rates, even in short-term follow-up, and with a recent long-term analysis by the MSKCC group documenting event-free survival of 6.1 months and overall survival of 12.9 months in patients treated with CAR-T therapy.173  Markers that distinguish a high risk of relapse vs long-term relapse-free survival after CAR-T therapy are actively being sought. Although greater persistence of CAR-T cells has been correlated with long-term survival in some studies (especially in studies of 4-1BB–containing CARs),137,138,144,155  this is not a universal finding, and, especially with CD28-containing CARs, long-term survival was most strongly correlated with the development of “deep” minimal residual disease–negative remission after CAR-T therapy and a lower disease burden at the start of treatment.173  Given the early stage of most of the CAR-T-cell studies, additional long-term studies and the discovery of surrogate markers of a sustained response to CAR-Ts are increasingly critical, as they will help inform decision-making about risk stratification post-CAR-T therapy. This risk stratification is of central importance in making decisions about what other therapies (if any) to offer patients who have been treated with CAR-Ts. Perhaps the most common of these postremission strategies is HCT. And, of note, although the MSKCC long-term data described similar outcomes for patients treated and not treated with HCT after CAR-T-cell therapy,173  this was a retrospective, not randomized-controlled analysis. However, these data and those from other trials suggest that carefully designed randomized trials of post–CAR-T-cell therapies (including determining whether post-CAR-T-cell HCT improves outcomes) are now warranted. These trials, as well as moving CAR-Ts earlier in the treatment paradigm for ALL represent the critical next phase of CD19-CAR-T clinical investigation. When discussing postremission therapeutic strategies for CAR-Ts, it is also critical to carefully consider the types of relapse that occur with the current generation of CAR-T products. In B-cell ALL, these relapses are both of CD19+ leukemia (indicating lack of long-term efficacy of the CARs) and of CD19 clones, indicating mutational escape as a pivotal event in disease relapse.111,112,137,138,144,152,154,155,173,175-177  The next generation of CARs is tackling both of these modes of treatment failure, and if successful, could further reduce the need for postremission therapies.

In this review, one of the oldest (HCT) and one of the newest (CAR-T) cellular therapies have been discussed, each of which plays a critical role in the treatment of patients with hematologic diseases. The reality of cellular therapeutics is that they have a narrow therapeutic window, with significant toxicities that often accompany their efficacy. Understanding the mechanisms driving both of these processes, and identifying surrogates to help optimize the risk-to-benefit ratio of these and other cellular therapies, will be critical to their successful implementation in patients with hematologic diseases.

L.S.K. was supported by National Institutes of Health, National Heart, Lung, and Blood Institute grant 1R01HL095791, and National Institute of Allergy and Infectious Diseases grants 2U19AI051731, 1UM1AI126617, U19AI117945, and R33AI116184.

Contribution: L.S.K. wrote the manuscript.

Conflict-of-interest disclosure: L.S.K. has received research funding from Juno Therapeutics, Bristol-Myers Squibb, Kymab Ltd, and Regeneron, and is a consultant for Bristol-Myers Squibb, Kymab, Regeneron, and Equilium.

Correspondence: Leslie S. Kean, The Ben Towne Center for Childhood Cancer Research, Seattle Children's Research Institute, Clinical Research Division, The Fred Hutchinson Cancer Research Center, and the Department of Pediatrics, University of Washington, 1100 Olive Way, Suite 100, Seattle, WA 98101; e-mail: leslie.kean@seattlechildrens.org.

1.
Sorror
ML
,
Maris
MB
,
Storb
R
, et al
.
Hematopoietic cell transplantation (HCT)-specific comorbidity index: a new tool for risk assessment before allogeneic HCT
.
Blood
.
2005
;
106
(
8
):
2912
-
2919
.
2.
Vaughn
JE
,
Storer
BE
,
Armand
P
, et al
.
Design and validation of an augmented hematopoietic cell transplantation-comorbidity index comprising pretransplant ferritin, albumin, and platelet count for prediction of outcomes after allogeneic transplantation
.
Biol Blood Marrow Transplant
.
2015
;
21
(
8
):
1418
-
1424
.
3.
Orozco
JJ
,
Kenoyer
A
,
Balkin
ER
, et al
.
Anti-CD45 radioimmunotherapy without TBI before transplantation facilitates persistent haploidentical donor engraftment
.
Blood
.
2016
;
127
(
3
):
352
-
359
.
4.
Mawad
R
,
Gooley
TA
,
Rajendran
JG
, et al
.
Radiolabeled anti-CD45 antibody with reduced-intensity conditioning and allogeneic transplantation for younger patients with advanced acute myeloid leukemia or myelodysplastic syndrome
.
Biol Blood Marrow Transplant
.
2014
;
20
(
9
):
1363
-
1368
.
5.
Nakamae
H
,
Wilbur
DS
,
Hamlin
DK
, et al
.
Biodistributions, myelosuppression, and toxicities in mice treated with an anti-CD45 antibody labeled with the alpha-emitting radionuclides bismuth-213 or astatine-211
.
Cancer Res
.
2009
;
69
(
6
):
2408
-
2415
.
6.
Chhabra
A
,
Ring
AM
,
Weiskopf
K
, et al
.
Hematopoietic stem cell transplantation in immunocompetent hosts without radiation or chemotherapy
.
Sci Transl Med
.
2016
;
8
(
351
):
351ra105
.
7.
Palchaudhuri
R
,
Saez
B
,
Hoggatt
J
, et al
.
Non-genotoxic conditioning for hematopoietic stem cell transplantation using a hematopoietic-cell-specific internalizing immunotoxin
.
Nat Biotechnol
.
2016
;
34
(
7
):
738
-
745
.
8.
Martin
PJ
,
Hansen
JA
,
Thomas
ED
.
Preincubation of donor bone marrow cells with a combination of murine monoclonal anti-T-cell antibodies without complement does not prevent graft-versus-host disease after allogeneic marrow transplantation
.
J Clin Immunol
.
1984
;
4
(
1
):
18
-
22
.
9.
Martin
PJ
,
Hansen
JA
.
Quantitative assays for detection of residual T cells of T-depleted human marrow
.
Blood
.
1985
;
65
(
5
):
1134
-
1140
.
10.
Martin
PJ
,
Hansen
JA
,
Storb
R
,
Thomas
ED
.
T cell depletion of donor marrow for prevention of acute graft-versus-host disease
.
Haematol Blood Transfus
.
1985
;
29
:
42
-
43
.
11.
Martin
PJ
,
Hansen
JA
,
Buckner
CD
, et al
.
Effects of in vitro depletion of T cells in HLA-identical allogeneic marrow grafts
.
Blood
.
1985
;
66
(
3
):
664
-
672
.
12.
O’Reilly
RJ
,
Collins
N
,
Dinsmore
R
, et al
.
Transplantation of HLA-mismatched marrow depleted of T-cells by lectin agglutination and E-rosette depletion
.
Tokai J Exp Clin Med
.
1985
;
10
(
2-3
):
99
-
107
.
13.
O’Reilly
RJ
,
Brochstein
J
,
Collins
N
, et al
.
Evaluation of HLA-haplotype disparate parental marrow grafts depleted of T lymphocytes by differential agglutination with a soybean lectin and E-rosette depletion for the treatment of severe combined immunodeficiency
.
Vox Sang
.
1986
;
51
(
suppl 2
):
81
-
86
.
14.
Wimperis
JZ
,
Brenner
MK
,
Drexler
HG
,
Hoffbrand
AV
,
Prentice
HG
.
Rapid recovery of helper activity following T cell depleted allogeneic marrow transplant
.
Clin Exp Immunol
.
1987
;
69
(
3
):
601
-
610
.
15.
Brenner
MK
,
Wimperis
JZ
,
Reittie
JE
, et al
.
Recovery of immunoglobulin isotypes following T-cell depleted allogeneic bone marrow transplantation
.
Br J Haematol
.
1986
;
64
(
1
):
125
-
132
.
16.
Parreira
A
,
Smith
J
,
Hows
JM
, et al
.
Immunological reconstitution after bone marrow transplant with Campath-1 treated bone marrow
.
Clin Exp Immunol
.
1987
;
67
(
1
):
142
-
150
.
17.
Apperley
JF
,
Mauro
FR
,
Goldman
JM
, et al
.
Bone marrow transplantation for chronic myeloid leukaemia in first chronic phase: importance of a graft-versus-leukaemia effect
.
Br J Haematol
.
1988
;
69
(
2
):
239
-
245
.
18.
Goldman
JM
,
Gale
RP
,
Horowitz
MM
, et al
.
Bone marrow transplantation for chronic myelogenous leukemia in chronic phase. Increased risk for relapse associated with T-cell depletion
.
Ann Intern Med
.
1988
;
108
(
6
):
806
-
814
.
19.
Martelli
MF
,
Aversa
F
.
Haploidentical transplants using ex vivo T-cell depletion
.
Semin Hematol
.
2016
;
53
(
4
):
252
-
256
.
20.
Marmont
AM
,
Horowitz
MM
,
Gale
RP
, et al
.
T-cell depletion of HLA-identical transplants in leukemia
.
Blood
.
1991
;
78
(
8
):
2120
-
2130
.
21.
Martin
PJ
,
Hansen
JA
,
Torok-Storb
B
, et al
.
Graft failure in patients receiving T cell-depleted HLA-identical allogeneic marrow transplants
.
Bone Marrow Transplant
.
1988
;
3
(
5
):
445
-
456
.
22.
Aversa
F
,
Terenzi
A
,
Carotti
A
, et al
.
Improved outcome with T-cell-depleted bone marrow transplantation for acute leukemia
.
J Clin Oncol
.
1999
;
17
(
5
):
1545
-
1550
.
23.
Aversa
F
,
Tabilio
A
,
Velardi
A
, et al
.
Treatment of high-risk acute leukemia with T-cell-depleted stem cells from related donors with one fully mismatched HLA haplotype
.
N Engl J Med
.
1998
;
339
(
17
):
1186
-
1193
.
24.
Aversa
F
,
Terenzi
A
,
Tabilio
A
, et al
.
Full haplotype-mismatched hematopoietic stem-cell transplantation: a phase II study in patients with acute leukemia at high risk of relapse
.
J Clin Oncol
.
2005
;
23
(
15
):
3447
-
3454
.
25.
Papadopoulos
EB
,
Carabasi
MH
,
Castro-Malaspina
H
, et al
.
T-cell-depleted allogeneic bone marrow transplantation as postremission therapy for acute myelogenous leukemia: freedom from relapse in the absence of graft-versus-host disease
.
Blood
.
1998
;
91
(
3
):
1083
-
1090
.
26.
Bayraktar
UD
,
de Lima
M
,
Saliba
RM
, et al
.
Ex vivo T cell-depleted versus unmodified allografts in patients with acute myeloid leukemia in first complete remission
.
Biol Blood Marrow Transplant
.
2013
;
19
(
6
):
898
-
903
.
27.
Jakubowski
AA
,
Small
TN
,
Young
JW
, et al
.
T cell depleted stem-cell transplantation for adults with hematologic malignancies: sustained engraftment of HLA-matched related donor grafts without the use of antithymocyte globulin
.
Blood
.
2007
;
110
(
13
):
4552
-
4559
.
28.
Handgretinger
R
.
New approaches to graft engineering for haploidentical bone marrow transplantation
.
Semin Oncol
.
2012
;
39
(
6
):
664
-
673
.
29.
Handgretinger
R
.
Negative depletion of CD3(+) and TcRαβ(+) T cells
.
Curr Opin Hematol
.
2012
;
19
(
6
):
434
-
439
.
30.
Carding
SR
,
Egan
PJ
.
Gammadelta T cells: functional plasticity and heterogeneity
.
Nat Rev Immunol
.
2002
;
2
(
5
):
336
-
345
.
31.
Locatelli
F
,
Bauquet
A
,
Palumbo
G
,
Moretta
F
,
Bertaina
A
.
Negative depletion of α/β+ T cells and of CD19+ B lymphocytes: a novel frontier to optimize the effect of innate immunity in HLA-mismatched hematopoietic stem cell transplantation
.
Immunol Lett
.
2013
;
155
(
1-2
):
21
-
23
.
32.
Bertaina
A
,
Merli
P
,
Rutella
S
, et al
.
HLA-haploidentical stem cell transplantation after removal of αβ+ T and B cells in children with nonmalignant disorders
.
Blood
.
2014
;
124
(
5
):
822
-
826
.
33.
Airoldi
I
,
Bertaina
A
,
Prigione
I
, et al
.
γδ T-cell reconstitution after HLA-haploidentical hematopoietic transplantation depleted of TCR-αβ+/CD19+ lymphocytes
.
Blood
.
2015
;
125
(
15
):
2349
-
2358
.
34.
Stern
M
,
Ruggeri
L
,
Mancusi
A
, et al
.
Survival after T cell-depleted haploidentical stem cell transplantation is improved using the mother as donor
.
Blood
.
2008
;
112
(
7
):
2990
-
2995
.
35.
Di Stasi
A
,
Tey
SK
,
Dotti
G
, et al
.
Inducible apoptosis as a safety switch for adoptive cell therapy
.
N Engl J Med
.
2011
;
365
(
18
):
1673
-
1683
.
36.
Zhou
X
,
Di Stasi
A
,
Tey
SK
, et al
.
Long-term outcome after haploidentical stem cell transplant and infusion of T cells expressing the inducible caspase 9 safety transgene
.
Blood
.
2014
;
123
(
25
):
3895
-
3905
.
37.
Zhou
X
,
Brenner
MK
.
Improving the safety of T-Cell therapies using an inducible caspase-9 gene
.
Exp Hematol
.
2016
;
44
(
11
):
1013
-
1019
.
38.
Tey
SK
,
Dotti
G
,
Rooney
CM
,
Heslop
HE
,
Brenner
MK
.
Inducible caspase 9 suicide gene to improve the safety of allodepleted T cells after haploidentical stem cell transplantation
.
Biol Blood Marrow Transplant
.
2007
;
13
(
8
):
913
-
924
.
39.
Straathof
KC
,
Pulè
MA
,
Yotnda
P
, et al
.
An inducible caspase 9 safety switch for T-cell therapy
.
Blood
.
2005
;
105
(
11
):
4247
-
4254
.
40.
Al Malki
MM
,
Horowitz
M
,
Handgretinger
R
, et al
.
Proceedings from the Second Haploidentical Stem Cell Transplantation Symposium-Haplo2014, San Francisco, California, December 4, 2014
.
Biol Blood Marrow Transplant
.
2016
;
22
(
4
):
594
-
604
.
41.
Zhou
X
,
Dotti
G
,
Krance
RA
, et al
.
Inducible caspase-9 suicide gene controls adverse effects from alloreplete T cells after haploidentical stem cell transplantation
.
Blood
.
2015
;
125
(
26
):
4103
-
4113
.
42.
Anderson
BE
,
McNiff
J
,
Yan
J
, et al
.
Memory CD4+ T cells do not induce graft-versus-host disease
.
J Clin Invest
.
2003
;
112
(
1
):
101
-
108
.
43.
Zheng
H
,
Matte-Martone
C
,
Jain
D
,
McNiff
J
,
Shlomchik
WD
.
Central memory CD8+ T cells induce graft-versus-host disease and mediate graft-versus-leukemia
.
J Immunol
.
2009
;
182
(
10
):
5938
-
5948
.
44.
Anderson
BE
,
Taylor
PA
,
McNiff
JM
, et al
.
Effects of donor T-cell trafficking and priming site on graft-versus-host disease induction by naive and memory phenotype CD4 T cells
.
Blood
.
2008
;
111
(
10
):
5242
-
5251
.
45.
Shook
DR
,
Triplett
BM
,
Eldridge
PW
,
Kang
G
,
Srinivasan
A
,
Leung
W
.
Haploidentical stem cell transplantation augmented by CD45RA negative lymphocytes provides rapid engraftment and excellent tolerability
.
Pediatr Blood Cancer
.
2015
;
62
(
4
):
666
-
673
.
46.
Triplett
BM
,
Shook
DR
,
Eldridge
P
, et al
.
Rapid memory T-cell reconstitution recapitulating CD45RA-depleted haploidentical transplant graft content in patients with hematologic malignancies
[published correction appears in Bone Marrow Transplant. 2015;50(7):1012].
Bone Marrow Transplant
.
2015
;
50
(
7
):
968
-
977
.
47.
Bleakley
M
,
Heimfeld
S
,
Loeb
KR
, et al
.
Outcomes of acute leukemia patients transplanted with naive T cell-depleted stem cell grafts
.
J Clin Invest
.
2015
;
125
(
7
):
2677
-
2689
.
48.
Bleakley
M
,
Heimfeld
S
,
Jones
LA
, et al
.
Engineering human peripheral blood stem cell grafts that are depleted of naïve T cells and retain functional pathogen-specific memory T cells
.
Biol Blood Marrow Transplant
.
2014
;
20
(
5
):
705
-
716
.
49.
Langston
AA
,
Prichard
JM
,
Muppidi
S
, et al
.
Favorable impact of pre-transplant ATG on outcomes of reduced-intensity hematopoietic cell transplants from partially mismatched unrelated donors
.
Bone Marrow Transplant
.
2014
;
49
(
2
):
185
-
189
.
50.
Suessmuth
Y
,
Mukherjee
R
,
Watkins
B
, et al
.
CMV reactivation drives posttransplant T-cell reconstitution and results in defects in the underlying TCRβ repertoire
.
Blood
.
2015
;
125
(
25
):
3835
-
3850
.
51.
Kanakry
CG
,
Coffey
DG
,
Towlerton
AM
, et al
.
Origin and evolution of the T cell repertoire after posttransplantation cyclophosphamide
.
JCI Insight
.
2016
;
1
(
5
).
52.
Goldberg
JD
,
Zheng
J
,
Ratan
R
, et al
.
Early recovery of T-cell function predicts improved survival after T-cell depleted allogeneic transplant
.
Leuk Lymphoma
.
2017
;
58
(
8
):
1859
-
1871
.
53.
Douek
DC
,
Vescio
RA
,
Betts
MR
, et al
.
Assessment of thymic output in adults after haematopoietic stem-cell transplantation and prediction of T-cell reconstitution
.
Lancet
.
2000
;
355
(
9218
):
1875
-
1881
.
54.
Sutherland
JS
,
Goldberg
GL
,
Hammett
MV
, et al
.
Activation of thymic regeneration in mice and humans following androgen blockade
.
J Immunol
.
2005
;
175
(
4
):
2741
-
2753
.
55.
Williams
KM
,
Moore
AR
,
Lucas
PJ
,
Wang
J
,
Bare
CV
,
Gress
RE
.
FLT3 ligand regulates thymic precursor cells and hematopoietic stem cells through interactions with CXCR4 and the marrow niche
.
Exp Hematol
.
2017
;
52
:
40
-
49
.
56.
Williams
KM
,
Lucas
PJ
,
Bare
CV
, et al
.
CCL25 increases thymopoiesis after androgen withdrawal
.
Blood
.
2008
;
112
(
8
):
3255
-
3263
.
57.
Duinhouwer
LE
,
Beije
N
,
van der Holt
B
, et al
.
Impaired thymopoiesis predicts for a high risk of severe infections after reduced intensity conditioning without anti-thymocyte globulin in double umbilical cord blood transplantation
[
published online ahead of print 9 February 2018
].
Bone Marrow Transplant
. doi:10.1038/s41409-018-0103-y.
58.
Velardi
E
,
Tsai
JJ
,
Radtke
S
, et al
.
Suppression of luteinizing hormone enhances HSC recovery after hematopoietic injury
.
Nat Med
.
2018
;
24
(
2
):
239
-
246
.
59.
Wertheimer
T
,
Velardi
E
,
Tsai
J
, et al
.
Production of BMP4 by endothelial cells is crucial for endogenous thymic regeneration
.
Sci Immunol
.
2018
;
3
(
19
).
60.
Hartwell
MJ
,
Özbek
U
,
Holler
E
, et al
.
An early-biomarker algorithm predicts lethal graft-versus-host disease and survival
.
JCI Insight
.
2017
;
2
(
3
):
e89798
.
61.
Levine
JE
,
Braun
TM
,
Harris
AC
, et al
;
Blood and Marrow Transplant Clinical Trials Network
.
A prognostic score for acute graft-versus-host disease based on biomarkers: a multicentre study
.
Lancet Haematol
.
2015
;
2
(
1
):
e21
-
e29
.
62.
Gatza
E
,
Braun
T
,
Levine
JE
, et al
.
Etanercept plus topical corticosteroids as initial therapy for grade one acute graft-versus-host disease after allogeneic hematopoietic cell transplantation
.
Biol Blood Marrow Transplant
.
2014
;
20
(
9
):
1426
-
1434
.
63.
Vander Lugt
MT
,
Braun
TM
,
Hanash
S
, et al
.
ST2 as a marker for risk of therapy-resistant graft-versus-host disease and death
.
N Engl J Med
.
2013
;
369
(
6
):
529
-
539
.
64.
Harris
AC
,
Ferrara
JL
,
Levine
JE
.
Advances in predicting acute GVHD
.
Br J Haematol
.
2013
;
160
(
3
):
288
-
302
.
65.
Harris
AC
,
Levine
JE
,
Ferrara
JL
.
Have we made progress in the treatment of GVHD?
Best Pract Res Clin Haematol
.
2012
;
25
(
4
):
473
-
478
.
66.
Levine
JE
,
Logan
BR
,
Wu
J
, et al
.
Acute graft-versus-host disease biomarkers measured during therapy can predict treatment outcomes: a Blood and Marrow Transplant Clinical Trials Network study
.
Blood
.
2012
;
119
(
16
):
3854
-
3860
.
67.
Harris
AC
,
Ferrara
JL
,
Braun
TM
, et al
.
Plasma biomarkers of lower gastrointestinal and liver acute GVHD
.
Blood
.
2012
;
119
(
12
):
2960
-
2963
.
68.
Ferrara
JL
,
Harris
AC
,
Greenson
JK
, et al
.
Regenerating islet-derived 3-alpha is a biomarker of gastrointestinal graft-versus-host disease
.
Blood
.
2011
;
118
(
25
):
6702
-
6708
.
69.
Paczesny
S
,
Braun
TM
,
Levine
JE
, et al
.
Elafin is a biomarker of graft-versus-host disease of the skin
.
Sci Transl Med
.
2010
;
2
(
13
):
13ra2
.
70.
Paczesny
S
,
Levine
JE
,
Braun
TM
,
Ferrara
JL
.
Plasma biomarkers in graft-versus-host disease: a new era?
Biol Blood Marrow Transplant
.
2009
;
15
(
suppl 1
):
33
-
38
.
71.
Paczesny
S
,
Krijanovski
OI
,
Braun
TM
, et al
.
A biomarker panel for acute graft-versus-host disease
.
Blood
.
2009
;
113
(
2
):
273
-
278
.
72.
Kitko
CL
,
Paczesny
S
,
Yanik
G
, et al
.
Plasma elevations of tumor necrosis factor-receptor-1 at day 7 postallogeneic transplant correlate with graft-versus-host disease severity and overall survival in pediatric patients
.
Biol Blood Marrow Transplant
.
2008
;
14
(
7
):
759
-
765
.
73.
Major-Monfried
H
,
Renteria
AS
,
Pawarode
A
, et al
.
MAGIC biomarkers predict long term outcomes for steroid-resistant acute GVHD
[
published online ahead of print 15 March 2018
].
Blood
. doi:10.1182/blood-2018-01-822957.
74.
MacMillan
ML
,
Robin
M
,
Harris
AC
, et al
.
A refined risk score for acute graft-versus-host disease that predicts response to initial therapy, survival, and transplant-related mortality
.
Biol Blood Marrow Transplant
.
2015
;
21
(
4
):
761
-
767
.
75.
Kitko
CL
,
Levine
JE
,
Storer
BE
, et al
.
Plasma CXCL9 elevations correlate with chronic GVHD diagnosis
.
Blood
.
2014
;
123
(
5
):
786
-
793
.
76.
Wolff
D
,
Greinix
H
,
Lee
SJ
, et al
.
Biomarkers in chronic graft-versus-host disease: quo vadis
[
published online ahead of print 24 January 2018
]?
Bone Marrow Transplant
. doi:10.1038/s41409-018-0092-x.
77.
Paczesny
S
,
Hakim
FT
,
Pidala
J
, et al
.
National Institutes of Health Consensus Development Project on Criteria for Clinical Trials in Chronic Graft-versus-Host Disease: III. The 2014 Biomarker Working Group Report
.
Biol Blood Marrow Transplant
.
2015
;
21
(
5
):
780
-
792
.
78.
Pidala
J
,
Sarwal
M
,
Roedder
S
,
Lee
SJ
.
Biologic markers of chronic GVHD
.
Bone Marrow Transplant
.
2014
;
49
(
3
):
324
-
331
.
79.
Pidala
J
,
Chai
X
,
Martin
P
, et al
.
Hand grip strength and 2-minute walk test in chronic graft-versus-host disease assessment: analysis from the Chronic GVHD Consortium
.
Biol Blood Marrow Transplant
.
2013
;
19
(
6
):
967
-
972
.
80.
Goldberg
JD
,
Giralt
S
.
Assessing response of therapy for acute and chronic graft-versus-host disease
.
Expert Rev Hematol
.
2013
;
6
(
1
):
103
-
107
.
81.
Lee
SJ
,
Flowers
ME
.
Recognizing and managing chronic graft-versus-host disease
.
Hematology Am Soc Hematol Educ Program
.
2008
;
2008
:
134
-
141
.
82.
Pavletic
SZ
,
Lee
SJ
,
Socie
G
,
Vogelsang
G
.
Chronic graft-versus-host disease: implications of the National Institutes of Health consensus development project on criteria for clinical trials
.
Bone Marrow Transplant
.
2006
;
38
(
10
):
645
-
651
.
83.
Schultz
KR
,
Miklos
DB
,
Fowler
D
, et al
.
Toward biomarkers for chronic graft-versus-host disease: National Institutes of Health Consensus Development Project on criteria for clinical trials in chronic graft-versus-host disease: III. Biomarker Working Group Report
.
Biol Blood Marrow Transplant
.
2006
;
12
(
2
):
126
-
137
.
84.
Pidala
J
,
Sigdel
TK
,
Wang
A
, et al
.
A combined biomarker and clinical panel for chronic graft versus host disease diagnosis
.
J Pathol Clin Res
.
2016
;
3
(
1
):
3
-
16
.
85.
Yu
J
,
Storer
BE
,
Kushekhar
K
, et al
.
Biomarker panel for chronic graft-versus-host disease
.
J Clin Oncol
.
2016
;
34
(
22
):
2583
-
2590
.
86.
Gartlan
KH
,
Bommiasamy
H
,
Paz
K
, et al
.
A critical role for donor-derived IL-22 in cutaneous chronic GVHD
.
Am J Transplant
.
2018
;
18
(
4
):
810
-
820
.
87.
Poe
JC
,
Jia
W
,
Su
H
, et al
.
An aberrant NOTCH2-BCR signaling axis in B cells from patients with chronic GVHD
.
Blood
.
2017
;
130
(
19
):
2131
-
2145
.
88.
Du
J
,
Paz
K
,
Flynn
R
, et al
.
Pirfenidone ameliorates murine chronic GVHD through inhibition of macrophage infiltration and TGF-β production
.
Blood
.
2017
;
129
(
18
):
2570
-
2580
.
89.
Cooke
KR
,
Luznik
L
,
Sarantopoulos
S
, et al
.
The biology of chronic graft-versus-host disease: a task force report from the National Institutes of Health consensus development project on criteria for clinical trials in chronic graft-versus-host disease
.
Biol Blood Marrow Transplant
.
2017
;
23
(
2
):
211
-
234
.
90.
Saha
A
,
O’Connor
RS
,
Thangavelu
G
, et al
.
Programmed death ligand-1 expression on donor T cells drives graft-versus-host disease lethality
.
J Clin Invest
.
2016
;
126
(
7
):
2642
-
2660
.
91.
Zeiser
R
,
Blazar
BR
.
Preclinical models of acute and chronic graft-versus-host disease: how predictive are they for a successful clinical translation?
Blood
.
2016
;
127
(
25
):
3117
-
3126
.
92.
Flynn
R
,
Paz
K
,
Du
J
, et al
.
Targeted Rho-associated kinase 2 inhibition suppresses murine and human chronic GVHD through a Stat3-dependent mechanism
.
Blood
.
2016
;
127
(
17
):
2144
-
2154
.
93.
Alexander
KA
,
Flynn
R
,
Lineburg
KE
, et al
.
CSF-1-dependent donor-derived macrophages mediate chronic graft-versus-host disease
.
J Clin Invest
.
2014
;
124
(
10
):
4266
-
4280
.
94.
Coghill
JM
,
Sarantopoulos
S
,
Moran
TP
,
Murphy
WJ
,
Blazar
BR
,
Serody
JS
.
Effector CD4+ T cells, the cytokines they generate, and GVHD: something old and something new
.
Blood
.
2011
;
117
(
12
):
3268
-
3276
.
95.
Gartlan
KH
,
Varelias
A
,
Koyama
M
, et al
.
Th17 plasticity and transition toward a pathogenic cytokine signature are regulated by cyclosporine after allogeneic SCT
.
Blood Adv
.
2017
;
1
(
6
):
341
-
351
.
96.
Varelias
A
,
Ormerod
KL
,
Bunting
MD
, et al
.
Acute graft-versus-host disease is regulated by an IL-17-sensitive microbiome
.
Blood
.
2017
;
129
(
15
):
2172
-
2185
.
97.
Le Texier
L
,
Lineburg
KE
,
Cao
B
, et al
.
Autophagy-dependent regulatory T cells are critical for the control of graft-versus-host disease
.
JCI Insight
.
2016
;
1
(
15
):
e86850
.
98.
Gartlan
KH
,
Markey
KA
,
Varelias
A
, et al
.
Tc17 cells are a proinflammatory, plastic lineage of pathogenic CD8+ T cells that induce GVHD without antileukemic effects
.
Blood
.
2015
;
126
(
13
):
1609
-
1620
.
99.
Koyama
M
,
Cheong
M
,
Markey
KA
, et al
.
Donor colonic CD103+ dendritic cells determine the severity of acute graft-versus-host disease
.
J Exp Med
.
2015
;
212
(
8
):
1303
-
1321
.
100.
Markey
KA
,
MacDonald
KP
,
Hill
GR
.
The biology of graft-versus-host disease: experimental systems instructing clinical practice
.
Blood
.
2014
;
124
(
3
):
354
-
362
.
101.
Koyama
M
,
Kuns
RD
,
Olver
SD
, et al
.
Promoting regulation via the inhibition of DNAM-1 after transplantation
.
Blood
.
2013
;
121
(
17
):
3511
-
3520
.
102.
Hill
GR
,
Kuns
RD
,
Raffelt
NC
, et al
.
SOCS3 regulates graft-versus-host disease
.
Blood
.
2010
;
116
(
2
):
287
-
296
.
103.
Tkachev
V
,
Furlan
SN
,
Watkins
B
, et al
.
Combined OX40L and mTOR blockade controls effector T cell activation while preserving Treg reconstitution after transplant
.
Sci Transl Med
.
2017
;
9
(
408
).
104.
Hippen
KL
,
Watkins
B
,
Tkachev
V
, et al
.
Preclinical testing of antihuman CD28 Fab’ antibody in a novel nonhuman primate small animal rodent model of xenogenic graft-versus-host disease
.
Transplantation
.
2016
;
100
(
12
):
2630
-
2639
.
105.
Furlan
SN
,
Watkins
B
,
Tkachev
V
, et al
.
Systems analysis uncovers inflammatory Th/Tc17-driven modules during acute GVHD in monkey and human T cells
.
Blood
.
2016
;
128
(
21
):
2568
-
2579
.
106.
Furlan
SN
,
Watkins
B
,
Tkachev
V
, et al
.
Transcriptome analysis of GVHD reveals aurora kinase A as a targetable pathway for disease prevention
.
Sci Transl Med
.
2015
;
7
(
315
):
315ra191
.
107.
Miller
WP
,
Srinivasan
S
,
Panoskaltsis-Mortari
A
, et al
.
GVHD after haploidentical transplantation: a novel, MHC-defined rhesus macaque model identifies CD28- CD8+ T cells as a reservoir of breakthrough T-cell proliferation during costimulation blockade and sirolimus-based immunosuppression
.
Blood
.
2010
;
116
(
24
):
5403
-
5418
.
108.
Grupp
SA
,
Kalos
M
,
Barrett
D
, et al
.
Chimeric antigen receptor-modified T cells for acute lymphoid leukemia
.
N Engl J Med
.
2013
;
368
(
16
):
1509
-
1518
.
109.
Ali
SA
,
Shi
V
,
Maric
I
, et al
.
T cells expressing an anti-B-cell maturation antigen chimeric antigen receptor cause remissions of multiple myeloma
.
Blood
.
2016
;
128
(
13
):
1688
-
1700
.
110.
Rosenberg
SA
,
Restifo
NP
.
Adoptive cell transfer as personalized immunotherapy for human cancer
.
Science
.
2015
;
348
(
6230
):
62
-
68
.
111.
Fry
TJ
,
Shah
NN
,
Orentas
RJ
, et al
.
CD22-targeted CAR T cells induce remission in B-ALL that is naive or resistant to CD19-targeted CAR immunotherapy
.
Nat Med
.
2018
;
24
(
1
):
20
-
28
.
112.
Fry
TJ
,
Mackall
CL
. T-cell adoptive immunotherapy for acute lymphoblastic leukemia.
Hematology Am Soc Hematol Educ Program
.
2013
;
2013
:
348
-
353
.
113.
Li
S
,
Tao
Z
,
Xu
Y
, et al
.
CD33 specific chimeric antigen receptor T cells with different costimulators showed potent anti-leukemia efficacy and different phenotype
.
Hum Gene Ther
.
2018
;
29
(
5
):
626
-
639
.
114.
Minagawa
K
,
Jamil
MO
,
Al-Obaidi
M
, et al
.
In vitro pre-clinical validation of suicide gene modified anti-CD33 redirected chimeric antigen receptor T-cells for acute myeloid leukemia
[published correction appears in PLoS One. 2017;12(2):e0172640].
PLoS One
.
2016
;
11
(
12
):
e0166891
.
115.
Cartellieri
M
,
Feldmann
A
,
Koristka
S
, et al
.
Switching CAR T cells on and off: a novel modular platform for retargeting of T cells to AML blasts
.
Blood Cancer J
.
2016
;
6
(
8
):
e458
.
116.
Kenderian
SS
,
Ruella
M
,
Shestova
O
, et al
.
CD33-specific chimeric antigen receptor T cells exhibit potent preclinical activity against human acute myeloid leukemia
.
Leukemia
.
2015
;
29
(
8
):
1637
-
1647
.
117.
Wang
QS
,
Wang
Y
,
Lv
HY
, et al
.
Treatment of CD33-directed chimeric antigen receptor-modified T cells in one patient with relapsed and refractory acute myeloid leukemia
.
Mol Ther
.
2015
;
23
(
1
):
184
-
191
.
118.
Pizzitola
I
,
Anjos-Afonso
F
,
Rouault-Pierre
K
, et al
.
Chimeric antigen receptors against CD33/CD123 antigens efficiently target primary acute myeloid leukemia cells in vivo
.
Leukemia
.
2014
;
28
(
8
):
1596
-
1605
.
119.
Lee
L
,
Draper
B
,
Chaplin
N
, et al
.
An APRIL-based chimeric antigen receptor for dual targeting of BCMA and TACI in multiple myeloma
.
Blood
.
2018
;
131
(
7
):
746
-
758
.
120.
Maciocia
PM
,
Wawrzyniecka
PA
,
Philip
B
, et al
.
Targeting the T cell receptor β-chain constant region for immunotherapy of T cell malignancies
.
Nat Med
.
2017
;
23
(
12
):
1416
-
1423
.
121.
Mamonkin
M
,
Rouce
RH
,
Tashiro
H
,
Brenner
MK
.
A T-cell-directed chimeric antigen receptor for the selective treatment of T-cell malignancies
.
Blood
.
2015
;
126
(
8
):
983
-
992
.
122.
Ramos
CA
,
Ballard
B
,
Zhang
H
, et al
.
Clinical and immunological responses after CD30-specific chimeric antigen receptor-redirected lymphocytes
.
J Clin Invest
.
2017
;
127
(
9
):
3462
-
3471
.
123.
Wang
CM
,
Wu
ZQ
,
Wang
Y
, et al
. Autologous T cells expressing CD30 chimeric antigen receptors for relapsed or refractory Hodgkin lymphoma: an open-label phase I trial.
Clin Cancer Res
.
2017
;
23
(
5
):
1156
-
1166
.
124.
Gomes-Silva
D
,
Srinivasan
M
,
Sharma
S
, et al
.
CD7-edited T cells expressing a CD7-specific CAR for the therapy of T-cell malignancies
.
Blood
.
2017
;
130
(
3
):
285
-
296
.
125.
Qasim
W
,
Zhan
H
,
Samarasinghe
S
, et al
.
Molecular remission of infant B-ALL after infusion of universal TALEN gene-edited CAR T cells
[published correction appears in Sci Transl Med. 2017;9(377)].
Sci Transl Med
.
2017
;
9
(
374
).
126.
Torikai
H
,
Reik
A
,
Liu
PQ
, et al
.
A foundation for universal T-cell based immunotherapy: T cells engineered to express a CD19-specific chimeric-antigen-receptor and eliminate expression of endogenous TCR
[published correction appears in Blood. 2015;126(22):2527].
Blood
.
2012
;
119
(
24
):
5697
-
5705
.
127.
Singh
N
,
Perazzelli
J
,
Grupp
SA
,
Barrett
DM
.
Early memory phenotypes drive T cell proliferation in patients with pediatric malignancies
.
Sci Transl Med
.
2016
;
8
(
320
):
320ra3
.
128.
Ceppi
F
,
Rivers
J
,
Annesley
C
, et al
.
Lymphocyte apheresis for chimeric antigen receptor T-cell manufacturing in children and young adults with leukemia and neuroblastoma
[
published online ahead of print 13 March 2018
].
Transfusion
. doi:10.1111/trf.14569.
129.
June
CH
,
O’Connor
RS
,
Kawalekar
OU
,
Ghassemi
S
,
Milone
MC
.
CAR T cell immunotherapy for human cancer
.
Science
.
2018
;
359
(
6382
):
1361
-
1365
.
130.
Srivastava
S
,
Riddell
SR
.
Chimeric antigen receptor T cell therapy: challenges to bench-to-bedside efficacy
.
J Immunol
.
2018
;
200
(
2
):
459
-
468
.
131.
Gomes-Silva
D
,
Ramos
CA
.
Cancer immunotherapy using CAR-T cells: from the research bench to the assembly line
.
Biotechnol J
.
2018
;
13
(
2
).
132.
Lim
WA
,
June
CH
.
The principles of engineering immune cells to treat cancer
.
Cell
.
2017
;
168
(
4
):
724
-
740
.
133.
Berger
C
,
Jensen
MC
,
Lansdorp
PM
,
Gough
M
,
Elliott
C
,
Riddell
SR
.
Adoptive transfer of effector CD8+ T cells derived from central memory cells establishes persistent T cell memory in primates
.
J Clin Invest
.
2008
;
118
(
1
):
294
-
305
.
134.
Gattinoni
L
,
Lugli
E
,
Ji
Y
, et al
.
A human memory T cell subset with stem cell-like properties
.
Nat Med
.
2011
;
17
(
10
):
1290
-
1297
.
135.
Wang
X
,
Berger
C
,
Wong
CW
,
Forman
SJ
,
Riddell
SR
,
Jensen
MC
.
Engraftment of human central memory-derived effector CD8+ T cells in immunodeficient mice
.
Blood
.
2011
;
117
(
6
):
1888
-
1898
.
136.
Sommermeyer
D
,
Hudecek
M
,
Kosasih
PL
, et al
.
Chimeric antigen receptor-modified T cells derived from defined CD8+ and CD4+ subsets confer superior antitumor reactivity in vivo
.
Leukemia
.
2016
;
30
(
2
):
492
-
500
.
137.
Turtle
CJ
,
Hanafi
LA
,
Berger
C
, et al
.
Immunotherapy of non-Hodgkin’s lymphoma with a defined ratio of CD8+ and CD4+ CD19-specific chimeric antigen receptor-modified T cells
.
Sci Transl Med
.
2016
;
8
(
355
):
355ra116
.
138.
Gardner
RA
,
Finney
O
,
Annesley
C
, et al
.
Intent-to-treat leukemia remission by CD19 CAR T cells of defined formulation and dose in children and young adults
.
Blood
.
2017
;
129
(
25
):
3322
-
3331
.
139.
Dummer
W
,
Niethammer
AG
,
Baccala
R
, et al
.
T cell homeostatic proliferation elicits effective antitumor autoimmunity
.
J Clin Invest
.
2002
;
110
(
2
):
185
-
192
.
140.
Goldrath
AW
,
Bevan
MJ
.
Selecting and maintaining a diverse T-cell repertoire
.
Nature
.
1999
;
402
(
6759
):
255
-
262
.
141.
Kaech
SM
,
Ahmed
R
.
Memory CD8+ T cell differentiation: initial antigen encounter triggers a developmental program in naïve cells
.
Nat Immunol
.
2001
;
2
(
5
):
415
-
422
.
142.
Gust
J
,
Hay
KA
,
Hanafi
LA
, et al
.
Endothelial activation and blood-brain barrier disruption in neurotoxicity after adoptive immunotherapy with CD19 CAR-T cells
.
Cancer Discov
.
2017
;
7
(
12
):
1404
-
1419
.
143.
Hay
KA
,
Hanafi
LA
,
Li
D
, et al
.
Kinetics and biomarkers of severe cytokine release syndrome after CD19 chimeric antigen receptor-modified T-cell therapy
.
Blood
.
2017
;
130
(
21
):
2295
-
2306
.
144.
Turtle
CJ
,
Hanafi
LA
,
Berger
C
, et al
.
CD19 CAR-T cells of defined CD4+:CD8+ composition in adult B cell ALL patients
.
J Clin Invest
.
2016
;
126
(
6
):
2123
-
2138
.
145.
Brentjens
RJ
,
Davila
ML
,
Riviere
I
, et al
.
CD19-targeted T cells rapidly induce molecular remissions in adults with chemotherapy-refractory acute lymphoblastic leukemia
.
Sci Transl Med
.
2013
;
5
(
177
):
177ra38
.
146.
Cruz
CR
,
Micklethwaite
KP
,
Savoldo
B
, et al
.
Infusion of donor-derived CD19-redirected virus-specific T cells for B-cell malignancies relapsed after allogeneic stem cell transplant: a phase 1 study
.
Blood
.
2013
;
122
(
17
):
2965
-
2973
.
147.
Garfall
AL
,
Maus
MV
,
Hwang
WT
, et al
.
Chimeric antigen receptor T cells against CD19 for multiple myeloma
.
N Engl J Med
.
2015
;
373
(
11
):
1040
-
1047
.
148.
Kebriaei
P
,
Singh
H
,
Huls
MH
, et al
.
Phase I trials using Sleeping Beauty to generate CD19-specific CAR T cells
.
J Clin Invest
.
2016
;
126
(
9
):
3363
-
3376
.
149.
Kochenderfer
JN
,
Dudley
ME
,
Carpenter
RO
, et al
.
Donor-derived CD19-targeted T cells cause regression of malignancy persisting after allogeneic hematopoietic stem cell transplantation
.
Blood
.
2013
;
122
(
25
):
4129
-
4139
.
150.
Kochenderfer
JN
,
Wilson
WH
,
Janik
JE
, et al
.
Eradication of B-lineage cells and regression of lymphoma in a patient treated with autologous T cells genetically engineered to recognize CD19
.
Blood
.
2010
;
116
(
20
):
4099
-
4102
.
151.
Lee
DW
,
Kochenderfer
JN
,
Stetler-Stevenson
M
, et al
.
T cells expressing CD19 chimeric antigen receptors for acute lymphoblastic leukaemia in children and young adults: a phase 1 dose-escalation trial
.
Lancet
.
2015
;
385
(
9967
):
517
-
528
.
152.
Maude
SL
,
Frey
N
,
Shaw
PA
, et al
.
Chimeric antigen receptor T cells for sustained remissions in leukemia
.
N Engl J Med
.
2014
;
371
(
16
):
1507
-
1517
.
153.
Porter
DL
,
Levine
BL
,
Kalos
M
,
Bagg
A
,
June
CH
.
Chimeric antigen receptor-modified T cells in chronic lymphoid leukemia
.
N Engl J Med
.
2011
;
365
(
8
):
725
-
733
.
154.
Turtle
CJ
,
Maloney
DG
.
Clinical trials of CD19-targeted CAR-modified T cell therapy; a complex and varied landscape
.
Expert Rev Hematol
.
2016
;
9
(
8
):
719
-
721
.
155.
Turtle
CJ
,
Riddell
SR
,
Maloney
DG
.
CD19-Targeted chimeric antigen receptor-modified T-cell immunotherapy for B-cell malignancies
.
Clin Pharmacol Ther
.
2016
;
100
(
3
):
252
-
258
.
156.
Neelapu
SS
,
Tummala
S
,
Kebriaei
P
, et al
.
Chimeric antigen receptor T-cell therapy - assessment and management of toxicities
.
Nat Rev Clin Oncol
.
2018
;
15
(
1
):
47
-
62
.
157.
Teachey
DT
,
Bishop
MR
,
Maloney
DG
,
Grupp
SA
.
Toxicity management after chimeric antigen receptor T cell therapy: one size does not fit ‘ALL’
.
Nat Rev Clin Oncol
.
2018
;
15
(
4
):
218
.
158.
Locke
FL
,
Neelapu
SS
,
Bartlett
NL
, et al
.
Phase 1 results of ZUMA-1: a multicenter study of KTE-C19 anti-CD19 CAR T cell therapy in refractory aggressive lymphoma
.
Mol Ther
.
2017
;
25
(
1
):
285
-
295
.
159.
Neelapu
SS
,
Locke
FL
,
Bartlett
NL
, et al
.
Axicabtagene ciloleucel CAR T-cell therapy in refractory large B-cell lymphoma
.
N Engl J Med
.
2017
;
377
(
26
):
2531
-
2544
.
160.
Neelapu
SS
.
An interim analysis of the ZUMA-1 study of KTE-C19 in refractory, aggressive non-Hodgkin lymphoma
.
Clin Adv Hematol Oncol
.
2017
;
15
(
2
):
117
-
120
.
161.
Teachey
DT
,
Lacey
SF
,
Shaw
PA
, et al
.
Identification of predictive biomarkers for cytokine release syndrome after chimeric antigen receptor T-cell therapy for acute lymphoblastic leukemia
.
Cancer Discov
.
2016
;
6
(
6
):
664
-
679
.
162.
Davila
ML
,
Riviere
I
,
Wang
X
, et al
.
Efficacy and toxicity management of 19-28z CAR T cell therapy in B cell acute lymphoblastic leukemia
.
Sci Transl Med
.
2014
;
6
(
224
):
224ra25
.
163.
Brudno
JN
,
Kochenderfer
JN
.
Toxicities of chimeric antigen receptor T cells: recognition and management
.
Blood
.
2016
;
127
(
26
):
3321
-
3330
.
164.
Chen
F
,
Teachey
DT
,
Pequignot
E
, et al
.
Measuring IL-6 and sIL-6R in serum from patients treated with tocilizumab and/or siltuximab following CAR T cell therapy
.
J Immunol Methods
.
2016
;
434
:
1
-
8
.
165.
Lee
DW
,
Gardner
R
,
Porter
DL
, et al
.
Current concepts in the diagnosis and management of cytokine release syndrome
[published correction appears in Blood. 2015;126(8):1048].
Blood
.
2014
;
124
(
2
):
188
-
195
.
166.
Maude
SL
,
Barrett
D
,
Teachey
DT
,
Grupp
SA
.
Managing cytokine release syndrome associated with novel T cell-engaging therapies
.
Cancer J
.
2014
;
20
(
2
):
119
-
122
.
167.
Kochenderfer
JN
,
Dudley
ME
,
Kassim
SH
, et al
.
Chemotherapy-refractory diffuse large B-cell lymphoma and indolent B-cell malignancies can be effectively treated with autologous T cells expressing an anti-CD19 chimeric antigen receptor
.
J Clin Oncol
.
2015
;
33
(
6
):
540
-
549
.
168.
Brahmer
JR
,
Lacchetti
C
,
Schneider
BJ
, et al
.
Management of immune-related adverse events in patients treated with immune checkpoint inhibitor therapy: American Society of Clinical Oncology clinical practice guideline [published online ahead of print 14 February 2018]
.
J Clin Oncol
. doi:10.1200/JCO.2017.77.6385.
169.
Schuster
SJ
,
Svoboda
J
,
Chong
EA
, et al
.
Chimeric antigen receptor T cells in refractory B-cell lymphomas
.
N Engl J Med
.
2017
;
377
(
26
):
2545
-
2554
.
170.
Hu
Y
,
Sun
J
,
Wu
Z
, et al
.
Predominant cerebral cytokine release syndrome in CD19-directed chimeric antigen receptor-modified T cell therapy
.
J Hematol Oncol
.
2016
;
9
(
1
):
70
.
171.
Taraseviciute
A
,
Tkachev
V
,
Ponce
R
, et al
.
Chimeric antigen receptor T cell-mediated neurotoxicity in non-human primates [published online ahead of print 21 March 2018]
.
Cancer Discov
. doi:10.1158/2159-8290.CD-17-1368.
172.
Pennell
CA
,
Barnum
JL
,
McDonald-Hyman
CS
, et al
.
Human CD19-targeted mouse T cells induce B cell aplasia and toxicity in human CD19 transgenic mice [published online ahead of print 6 April 2018]
.
Mol Ther
. doi:10.1016/j.ymthe.2018.04.006.
173.
Park
JH
,
Rivière
I
,
Gonen
M
, et al
.
Long-term follow-up of CD19 CAR therapy in acute lymphoblastic leukemia
.
N Engl J Med
.
2018
;
378
(
5
):
449
-
459
.
174.
Brentjens
RJ
,
Rivière
I
,
Park
JH
, et al
.
Safety and persistence of adoptively transferred autologous CD19-targeted T cells in patients with relapsed or chemotherapy refractory B-cell leukemias
.
Blood
.
2011
;
118
(
18
):
4817
-
4828
.
175.
Gardner
R
,
Wu
D
,
Cherian
S
, et al
.
Acquisition of a CD19-negative myeloid phenotype allows immune escape of MLL-rearranged B-ALL from CD19 CAR-T-cell therapy
.
Blood
.
2016
;
127
(
20
):
2406
-
2410
.
176.
Park
JH
,
Geyer
MB
,
Brentjens
RJ
.
CD19-targeted CAR T-cell therapeutics for hematologic malignancies: interpreting clinical outcomes to date
.
Blood
.
2016
;
127
(
26
):
3312
-
3320
.
177.
Maude
SL
,
Laetsch
TW
,
Buechner
J
, et al
.
Tisagenlecleucel in children and young adults with B-cell lymphoblastic leukemia
.
N Engl J Med
.
2018
;
378
(
5
):
439
-
448
.
Sign in via your Institution