Back to Journals » Stem Cells and Cloning: Advances and Applications » Volume 13

Role of Stem-Cell Transplantation in Leukemia Treatment

Authors Dessie G , Derbew Molla M , Shibabaw T , Ayelign B 

Received 15 May 2020

Accepted for publication 25 July 2020

Published 10 August 2020 Volume 2020:13 Pages 67—77

DOI https://doi.org/10.2147/SCCAA.S262880

Checked for plagiarism Yes

Review by Single anonymous peer review

Peer reviewer comments 2

Editor who approved publication: Dr Bernard Binetruy



Gashaw Dessie,1 Meseret Derbew Molla,1 Tewodros Shibabaw,1 Birhanu Ayelign2

1Department of Biochemistry, School of Medicine, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia; 2Department of Immunology and Molecular Biology, School of Biomedical and Laboratory, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia

Correspondence: Gashaw Dessie
Department of Biochemistry, School of Medicine, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
Tel +251-97-515-2796
Email [email protected]

Abstract: Stem cells (SCs) play a major role in advanced fields of regenerative medicine and other research areas. They are involved in the regeneration of damaged tissue or cells, due to their self-renewal characteristics. Tissue or cells can be damaged through a variety of diseases, including hematologic and nonhematologic malignancies. In regard to this, stem-cell transplantation is a cellular therapeutic approach to restore those impaired cells, tissue, or organs. SCs have a therapeutic potential in the application of stem-cell transplantation. Research has been focused mainly on the application of hematopoietic SCs for transplantation. Cord blood cells and human leukocyte antigen–haploidentical donors are considered optional sources of hematopoietic stem–cell transplantation. On the other hand, pluripotent embryonic SCs and induced pluripotent SCs hold promise for advancement of stem-cell transplantation. In addition, nonhematopoietic mesenchymal SCs play their own significant role as a functional bone-marrow niche and in the management of graft-vs-host disease effects during the posttransplantation process. In this review, the role of different types of SCs is presented with regard to their application in SC transplantation. In addition to this, the therapeutic value of autologous and allogeneic hematopoietic stem–cell transplantation is assessed with respect to different types of leukemia. Highly advanced and progressive scientific research has focused on the application of stem-cell transplantation on specific leukemia types. We evaluated and compared the therapeutic potential of SC transplantation with various forms of leukemia. This review aimed to focus on the application of SCs in the treatment of leukemia.

Keywords: stem cell, leukemia, transplantation

Introduction

Stem cells (SCs) are undifferentiated cells that can be differentiated into other types of cell andalso have the potential to proliferate and self-renew to producenew SCs. In mammals, there are two broad type of SC. Embryonic SCs (ESCs) are present in the early life of the embryo and isolated from the inner cell massor morula of the blastocyst (future germ layer, such as endoderm, ectoderm, or mesoderm of the embryo).14 The surrounding section of the morula is known as the trophoblast, which can develop to the future placenta. Adult SCs (ASCs) are found in various tissue types of developed mammals.5 ASCs are useful for tissue regeneration and repair after severe injuries.1,6

SC populations may behave abnormally or be altered by genetic or environmental factor, resulting in the development of cancer. Leukemia comprises a group of hematologic disorders that usually begin in the bone marrow and resultin a high number of abnormal blood cells. It is the result of deregulation of normal hematopoietic SC (HSC) development by genetic mutation that produces a cell population known as leukemic SCs (LSCs). The generation of blood cells depends on the regulation of differentiation and proliferation characteristics of HSCs.7 Deregulated differentiation and proliferation activity of HSCs, including chromosomal translocation and somatic mutation, leads to different hematologic disorders. There are four major abnormalities identified under LSCs: such as acute myeloid leukemia (AML), acute lymphoblastic leukemia (ALL),8 chronic LL (CLL) and chronic ML (CML).4 Leukemia and lymphoma (Hodgkin’s lymphoma [HL] and non-HL [NHL]) are the two major types of blood cancers that result from uncontrolled proliferation of white blood cells, and were the first to be treated clinically using HSC transplantation (HSCT).1,–9-11 In addition, HSCT is used as a therapeutic option for many nonhematopoietic malignancies, aplastic anemia, and certain inherited disorders like severe thalassemia, sickle-cell disease, and other inherited metabolic disorders. Historically, HSCs were obtained only from bone marrow, but are now mostly harvested from peripheral blood after mobilization through administration of hemaTtopoietic growth factor and from the umbilical cord blood (UCB) of newborns.4,9

SC-based therapies become the major concern of researchers after the first effective bone-marrow transplant in 1968.12 Globally, food and drug administrations design regulations on the application of SC therapies. An increase in scientific knowledge of cell-differentiation pathways has promoted the application of SC therapy.12 Since the application of SC therapy emerged as a new insight into cellular therapeutic potential, food and drug administrations have continuously driven awareness and designed regulation with regard to SC therapies. SCs serve as a novel cellular therapeutic approach in the field of regenerative medicine to treat various disorders.13 In addition to renewing and proliferating themselves, they are capable of differentiation to specialized functional cells.14 This enables them to substitute various injured cells, such as cardiomyocytes, fibroblasts, and endothelial cells.15 In addition, regenerative medicine has significant therapeutic potential through the application of SCT to restore impaired blood cells.16

HSCT has broad application in treating different malignant and nonmalignant hematologic disorders. Researchers have noted that >40,000 HSCTs are performed every year to treat these disorders.17 In this context, autologous SCT (auto-SCT) and allogeneic SCT (allo-SCT) are the best known and most applicable.18 There are SC types that have the capability of being the source for SCTs. Bone-marrow SCs are the major sources for treating hematologic and nonhematologic disorders.19 Similarly, peripheral blood CD34+ cell have hematopoiesis potential for HSCT.20 With respect to recent scientific advancement,HSCs are generated from pluripotent ESCs that require the transition state from endothelial to hematopoietic progenitor cells to resolve HLA-mismatched problem.21 The recent investigation done by Serap et al (2019) and his colleagues hypothesized that achievement of effective HSCT may also associate with non-hematopoietic progenitor cells, very small embryonic-like SCs (VSELSCs).22 They differentiate into HSCs in vitro.23 With specific forward reprogramming protocols, induced pluripotent SCs (iPSCs) have therapeutic potential to generate hemato-endothelial progenitor (HEP) cells.

Co-administration of chemotherapy along with auto-SCT leads to a decrease in the level of regulatory T-cells. In response to the dysregulated immune system, biological characteristics of mesenchymal SCs (MSCs) contribute to hematopoietic reconstitution and an efficient HSC engraftment.24,25 On the other hand, bone marrow derived MSCs are other components of hematopoietic niche.26 Therefore, this review assessed different types of SCs that are utilized as the source and as support of SC transplantation. In addition, we also summarized the role of allogeneic and auto-SCT in the treatment of various types of leukemia.

Generation of Hematopoietic Stem Cells from Human Embryonic Stem Cells

The involvement of ESCs is the new therapeutic insights having a regenerative potential to restore impaired tissue or cells.27 ESCs are the source of SCs for cellular transplantation therapies; however, they may also lead to uncontrolled cell proliferation which also results in the development of cancers.28 The challenges of using these cells are their characteristic features of chromosomal abnormality and mutation during in vitro.29 Regard to this, c-MYC oncogene may be expressed that results in cancer cells than their cellular therapeutic significant.29 They require a safety concern due to their teratoma formation.30 Although they have teratoma effect, ESCs have a significant role in the transplantation process.28 Human ESCs (hESCs) serve as the source of development of cellular lineages through signaling pathways.13 Recently, protocols have been on the way to be designed to generate HSCs from pluripotent ESCs in vitro. The generation of HSCs from those pluripotent ESCs requires a transition from endothelial to hematopoietic progenitor cells to resolve HLAmismatching.21 The hematopoietic transcription factor Runx1 promotes the commitment of hematopoietic cellular lineages by activating the expression of Runx1a. NOTCH signaling enhances the transition state, while the TGFβ-signaling pathway inhibit it.31 Recently, generation of HSCs was achieved by Wang et al from hESCs andhumaniPSCs (Figure 1). The commitment stages that had been examined by those scientists confirmed the synthesis of hematopoietic cells from hESCs.32 In support of this, recently the ESC gene SLL4was identified and used as a therapeutic target for leukemia. Because of its importance in the ESC fate, SALL4 expression need to be reactivated during the reprogramming process of mouse embryonic fibroblasts to be converted into iPSCs. Under normal condition, SALL4 is expressed highly in CD34+CD38 HSCs and llittle in CD34+CD38 + hematopoietic progenitor cells. Therefore, the main application behind this ESC gene product is as key player in hematopoietic differentiation. Consequently, downregulation of this gene could be considered a therapeutic option for leukemia.33

Figure 1 Role of different types of SCs in SC transplantation. MSCs were the nonhematopoietic source utilized to reduce GVHD (reduce risk of graft failure by secreting soluble factors with anti-inflammatory properties), efficient HSCs support to engraftment of transplant, hematologic reconstitution, and to improve the HSCT outcome. HSCs can be generated from the hematoendothelial transition process from HESCs to HiPSCs, and commonly from bone-marrow SCs, PBSCs, and umbilical cord blood. The pluripotent potential of VSELSCs also enables to generate HSCs. Abbreviations: GVHD, graft-vs-host disease; HESCs, human embryonic SCs; HSCs, hematopoietic SCs; HSCT, hematopoietic SC transplantation; HiPSCs, human induced pluripotent SCs; MSCs, mesenchymal SCs; PBSC, peripheral blood SC; VSELSCs, very small embryonic-like SCs.

Generation of Hematopoietic Progenitor Cells from Induced Pluripotent Stem Cells

iPSCs were introduced as an alternative SC-based therapy method in 2006, by Takahashi and Yamanaka.34 Reprogramming of SCs through the integration of viruses with these cells induces differentiation capability in various tissue types.35 These are pSCs, which are generated from adult somatic cells through in vitro experimental investigation.36 They are synthesized in vitro by reprogramming mature mouse fibroblast cells through epigenetic modification.34 In human beings, production of iPSCs was started through the introduction of four genes — SOX2, MYC, OCT4, and KLF4 — into matured somatic fibroblasts37 and other human somatic cells.38 The genes are induced in these cells through the encoded retrovirus.39 The ability of iPSCs to expand into multicellular lineages enables them to be a potential SC-therapy method. Various types of patient-specific SCs have been synthesized from their expansion process in vitro.40 Research has revealed their cellular therapeutic significance in various hematologic malignancies, such as CML, MDS, AML,22 and BCR-ABL−myeloproliferative neoplasms.41 Donor blood cells are reprogrammed to iPSCs to generate patient-specific SCs.40 With specific forward-reprogramming protocols, iPSCs have the therapeutic potential to generate hematoendothelial progenitor cells. Lange et al demonstrate the possible generation of hematopoietic progenitor cells by combinatorial expression of transcription factors SCL, LMO2, GATA2, and ETV242 (Figure 1). Moreover, researchers have been trying to generate hematopoietic progenitor cells from PSCs. Shan et al described possible strategies for generation of HSCs from human mesenchymal cells with hematopoietic potential (Figure 1). They revealed the derivation or generation of hematopoietic progenitor cells from mouse PSCs using in vitro induction methods. Therefore, iPSCs can be have possible therapeutic potential in SCT; however, they present safety concerns, due to their teratoma formation.30 Allogeneic transplantation of bone marrow or umbilical cord reveals rejection, due to the effect of graft-vs-host disease (GVHD) and disease relapse, which restricts its applicability. In cases of auto-HSCT, there is no risk of rejection, but there remain leukemic cells that induce disease relapse. Collectively, these disadvantages of bone-marrow HSCT mandate alternative sources of HSCs aiming to reduce GVHD, disease relapse, and bone marrow–failure syndrome. Considering this, iPSCs represent a suitable source to generate HSCs in vitro with limited immunogenicity.43 These have a major advantage over bone-marrow and cord types, since their autologous transplantation from iPSCs does not induce GVHD.44

Very Small Embryonic-Like Stem Cells

Bhartiya et al characterized VSELSCs as the “true SCs” and the subset of different SC population, such as HSCs, ovarian SCs and MSCs. They express the OCT4A antigenic marker in their nucleus.30 The pluripotency features of VSELSCs enhance their expansion in vitro using the pyrimidoindole-derivative molecule UM171,45 and in turn are utilized for expansion of CD34+ HSCs.46 VSELSCs are involved in homeostatic processes, because they are found in quiescent stage, and later they differentiate into ASCs. They differentiate into HSCs in vitro.23 VSELSCs can be generated from primordial germ cells and undergo further differentiation into HSCs47 (Figure 1). Bone marrow–derived VSELSCs may not have features characteristic of hematopoietic progenitor SCs, but they can retain hematopoietic features through external-stress growth factors.48 The transcriptional factors Oct4A), Nanog, and Rex1 are found in VSELSCs, but they are not expressed in HSCs.22 Treatment of immunocompromised ALL8 patients with granulocyte colony–stimulating factorincreases mobilization of VSELSCs to the peripheral circulation.49 Dissemination of VSELSCs to the circulation promotes the regeneration of tissue.49 A recent investigation done by Serap et al hypothesized that achievement of effectiveHSCT may be associated with nonhematopoietic progenitor cells — VSELSCs.22 The expression of transcription factors and pluripotent markers may contribute to their therapeutic potential in SC transplantation. Demonstrations on immunocompromised mice have shown that VSELSCs have a lower teratoma effect.47 Similarly, an investigation done on animal models showed that they have the capability to differentiate into HSCs.46

Potential Effect of Cotransplantation of Mesenchymal Stem Cells to Treat Leukemia

Bone marrow–derived MSCs are important to regenerate injured tissue.50 Recently, MSCs have served as a new cellular therapy method in the field of regenerative medicine.13 They inhibit cancer-cell proliferation through secretion and inhibition of Dkk1- and Wnt-signaling pathways, respectively.51 Besides this, MSCs alter the immune system to regenerate damaged tissue and decrease inflammation.52 GVHD is one of the complications of both auto-SCT and allo-SCT during treatment.53 This posttransplantation complication is associated with immunologic intolerance.53 Indeed, MSCs have been shown to support the engraftment of autologously or allogeneically transplanted HSCs by secreting soluble factors or immunomodulators, such as TGFβ1 and HGF which inhibit the proliferation of CD4+ TH1, TH17, CD8+ T, and natural-killer cells, leading to prevention of GVHD.6,24,26 Therefore, GVHD that occurs after HSCT can be treated by coinfusion with MSCs.54 Bone marrow–derived MSCs are components of the hematopoietic niche. Additionally, they have the capability to regulate the hematopoiesis process through interactionand communicating with HSCs and progenitor cells55 (Figure 1).

Umbilical Cord Blood Cells and Haploidentical Transplantation

Donor availability is a very important issue, particularly in patients from ethnic minorities. A haploidentical donor and CB allow allo-HSCT in the majority of transplant-eligible patients.UCB is a well-established cellular product source for hematopoietic reconstitution and transplantation.37 It is derived from fetal tissue and acts as a potential source of progenitor SCs to synthesize matured HSCs16 (Figure 1). The lower complication rate of GVHD and less stringent HLA-matching requirements make it a valuable source of HSCs.56 It is more highly enriched with HSCs/progenitor cells than peripheral blood with regard to colony-forming unitgranulocyte/macrophage progenitors and CD34+-cell content.57

The effect of HLA mismatching is less severe in mismatched UCB transplantation than unrelated peripheral and bone marrow–blood transplantation;58 therefore, higher numbers of mismatched donors may donate to save lives. Compatibility at the DRB1-allele and HLA-A and -B antigen level is better for UCB transplantation to be selected traditionally without consideration of HLA-C.59 UCB has significance for allo-HSCT transplantation, because it requires lower HLA matching than for unrelated donors.59 In AML, unrelated CB transplantation has failed, due to nonrelapse mortality.60 However, the cost of CB delaying engraftment and risk of infection are still challenges in its application for hematologic diseases, including leukemia.61,62

In cases of rapid requirement of allograft and absence of an HLA-matched donor, HLA-haploidentical SC transplantation is considered a therapeutic option.63 Peripheral and bone-marrow SCs can be donated from these family members if they have one common haplotype.64 HLA-haploidentical cells are considered an optional source for HSCT.65 In haploidentical transplantation, the graft contains lower of T-cell content to diminish GVHD.66 Outcomes of haploidentical HSCT may be affected by innate immune cells like T cells and natural-killer cells.67 In high-risk acute leukemia, the applicabilion of HLA-haploidentical HSCT is elevated.65 However, outcomes of nonrelapse mortality and GVHD may be increased from haploidentical HSCT with higher HLA mismatching including from partially related donors, as the content of T-cell is replete.68

Bone-Marrow Stem Cells as Source for Stem-Cell Transplantation

A soft, gelatinous tissue, bone marrow is used as the source of peripheral HSCs.69 Researchers have argued that both bone marrow and peripheral blood are major sources of SCs. SCASCs generated from bone marrow are known as bone-marrow SCs,37 having clinical significance in restoring damaged cardiac tissue through gene therapy.70 Also, they can be a potential source for auto-HSCT..37 There is an improvement in GVHD in patients with bone-marrow SC transplantation compared to peripheral blood SCs (PBSCs).19 Bone marrow–SC transplantation is utilized in various hematologic malignancies, such as AML, ALL, and CML. The use of bone-marrow transplantation from compatible donors is the most effective treatment for CML.71 Allogenic bone-marrow transplantation is an effective alternative treatment option for patients who are resistant to chemoradiation therapy and have a higher probability of relapse.72 The physician removes marrow from the donor’s hip bone using surgical procedures, including anesthesia, sterile needles, and syringes, and replaces the donated bone marrow within 4–6 weeks. As the level of T cell compare in both bone-marrow transplantation and PBSCs, the concentration of T cells is reduced in bone-marrow transplantation.19

Peripheral Blood Stem Cells as Source for Stem-Cell Transplantation

Recent SC-transplantation protocols state that mobilization of HSCs from bone marrow to peripheral blood is an effective treatment method in the majority of transplanted patients.73 Although bone marrow is major source of SCs, a hematopoietic growth factor found in PBSCs showed that these are also another possible source of SCs.74 PBSCs from bone marrow are a valuable source in restoring hematologic disorders.69 The potential effect of PBSCs depends on hematopoietic development and enhancement of immunologic profiles, and hence they are a valuable source of HSCs to treat hematologic disorders. Peripheral blood CD34+ cells have hematopoietic potential for SCT.20 Javarappa et alpurified hematopoietic progenitor cells from CD4+ peripheral blood cellsafter which the cells differentiated into megakaryocytes and myeloid-lineage cells75 (Figure 1). PBSCs serve as a valuable SC source if mobilization is supported by granulocyte colony–stimulating factor.19 They are applicable in autolo-SCT in the treatment of multiple myeloma.76 The utilization of peripheral SCs as a source of SCs may induce the occurrence of GVHD.77 Even if they have such effects, the immune system has been enhanced, due to elevation of T-cell secretion. On the contrary, the elevation of T cells may also cause GVHD development;19 however, PBSC collection in children may expose them to metabolic complications, including hypocalcemia and hypoglycemia.78

Hematopoietic Stem-Cell Abnormalities in Leukemia

The tight control in proliferation and differentiation of HSCs has significant value for the synthesis of blood cells.7 Multipotent HSCs are responsible for cell division and proliferation.79 Somatic mutation of T cells during DNA methylation and posttransplantation alteration are risk factors for ALL.8,80 CML is a hematologic disorder induced by reverse chromosomal translocation on t(9;22)(q34;q11)81 and BCRABL oncogene effects on proliferative myelogenous cells.82 Mutated gene BCRABL, has a tyrosine-kinase effect and induces the release of highly proliferative myelogenous cells from bone marrow.81 The MYC gene is another oncogene that induces gene expression and has a proliferative effect on hematopoietic progenitor cells.83 In addition to this gene, BCL2 is another mutated gene that inhibits programmed cell death. As such, cancerous cells proceed with their continued proliferation and leukemic cells are released from the tissue where they were generated.84 Hitzler et al reported that a mutation of the GATA1 gene in acute megakaryoblastic leukemia affects hematopoietic transcriptional factor. On the other hand, chromosomal translocation of t(7;11)(p15;p15) HSCs lead to the integration of genes, including HOXA9 and NUP98, which also leads to distortion in the transcriptional process of hematopoietic precursor cells.85 Aberration of the transcriptional process in these cells induces abnormal cell proliferation, which may lead to AML.85 Overproliferation of lymphoblasts within bone marrow can also result in the pathogenesis of ALL.8,49

Hematopoietic Stem-Cell Transplantation to Treat Leukemia

Emphasis on the eradication of hematologic malignancies has shifted from cytotoxic chemotherapy to donors’ immune cells.86 HSCT is utilized by 20,000 people in the US every year.87 It is applicable in treating patients with rare diseases, such as AML,22 ALL,8 CML, Burkitt’s lymphoma, HL, and NHL,11 and other hematologic malignancies.88 Although it serves as an alternative treatment method, HSCT still has a relapse risk among 40%–80% of recipients.89 Both auto-HSCT and allo-HSCTare the main alternative cellular therapeutic methods to treat leukemia. Auto-HSCT is the appropriate and applicable therapeutic option for multiple myeloma1,18 and HL.11 Charles et al explained that auto-HSCT was more frequently utilized by European and North American countries than allo-HSCT to treat myeloma. A lower mortality rate for myeloma is seen with auto-HSCT. Auto-HSCT is an established treatment approach if myeloma is at an acute stage, but for older patients it requires extra improvement.90 The occurrence of GVHD among myeloma patients who undergo allo-HSCT is 50% compared to 5%–20% of occurrence of auto-SCTpatients91 (Figure 2). As such, fewer GVHD effects have been seen in auto-SCT n treating multiple myeloma and HL.11 Furthermore, in HIV-related lymphoma, auto-HSCT is considered an applicable therapeutic option in both relapsed HL1 and relapsed NHL patients.18,92

Figure 2 Comparison of allogeneic and autologous stem-cell transplantation with hematologic disorders. Autologous stem-cell transplantation has been utilized as a treatment protocol to treat MM and HL, due to its initial response, low relapse sensitivity, and positive positron-emission tomography (+PET). Patients at higher risk or progress of AML are treated with allo-HSCT. Chronic phase 1 (CP1), TKI intolerance, and blast crisis enables allo-HSCT to be a standard treatment option for the treatment of CML. Allo-HSCT is also a treatment option for NHL patients presenting with complete remission 1 and 2 (CR1 and CR2) indications and also relapse after auto-HSCT. Although they have graft-vs-leukemic toxic effects, they are a significant alternative cell-based therapy to treat hematologic malignancies. Abbreviations: ALL, acute lymphocytic leukemia; AML, acute myeloid leukemia; CML, chronic myeloid leukaemia; HL, Hodgkin’s lymphoma; MM, multiple myeloma; NHL, non-HL.

On the other hand, allo-HSCT is a curative treatment approach for severe AML93 It has been confirmed that hematologic toxicity is lower in these recipient patients. Allo-HSCT has also been used as a treatment option for acute lymphoid leukemia and multiple myeloma.1,23,94 Though alternative treatments remain undefined, it is a valuable treatment tool for hematologic malignancies. Reduced-intensity conditioning after allo-HSCT has been seen in Spain.95 The toxic effect of allo-HSCT is associated with graft-vs-leukemia reactions. Chronic myelogenous leukemia patients show lower relapse rate than other allogeneically transplanted leukemia patients.96 The therapeutic landscape of CML has shifted dramatically with developments tyrosine-kinase inhibitors (TKIs), which target the BCR–ABL1 hybrid oncoprotein and block the constitutive activity of tyrosine kinase. The course of CML is typically triphasic, with an early indolent chronic phase (CP), followed by an accelerated phase and a blast (crisis phase (BP).97,98 For selection of appropriate TKIs, of CML patients should be tested for BCR–ABL1 kinase–domain mutation (mutation profile), disease phase, and patient comorbidities. For example, if the patient has such mutations as Y253H, E255K/V, or F359C, physicians recommend dasatinib or bosutinib as TKI. On the other hand, if patients are in an advanced disease phase (BP) or CML-CP (with T315I mutation), third-generation ponatinib is preferred over imatinib.99103 However, allo-HSCT remains a therapeutic option for patients in CML-CP whose CML has progressed after at least two TKIs and after trialing ponatinib therapy (for T315I mutation) to reduce the CML burden, and for the effectiveness of the transplantation.99,100,102 An improvement in immunologic tolerance and lowered GVHD effect mean allo-HSCT is the only curative treatment option for CML-BP104 (Figure 2). Similarly to CML, highly complicated and severe AML is effectively treated with allo-HSCT.22 Complications of AML may lead to higher mortality and morbidity rates, which may be due to chronic GVHD among patients >50 years old.105 Pediatric ALL patients presenting with indications of higher relapse risk are treated (10% of treatment) with allo-HSCT.106

ALL patients who develop high relapse risk are indications for treatment with allo-HSCT.107 Allo-HSCT is a standard treatment method for ALL patients who are at higher risk.108 The use of allo-HSCT has lower toxicity in young patients.86 Allo-HSCT has lower relapse risk than auto-HSCT in multiple myeloma.18 Graft-vs-tumor reactions in hematologic malignancies depend on the donor’s T cells and donor lymphocyte infusions. The decision to perform allo-HSCT depends mainly on reduced intensity conditioning.109 Researchers haverecommended that the use of allo-HSCT should depend on strong clinical data; however, 28%–49% of allo-HSCT patients develop relapse risks for disease.110 Moreover, allo-HSCT has been widely applied as a therapeutic option in both HL and NHL.11

Conclusion

SCs play a major role in cell-based therapy to treat both hematologic and nonhematologic malignant disorders. They are mainly involved in the application of transplantation. Adult SCs (bone-marrow SCs), PBSCs, and UCB are the major potential sources of HSCs used during SC transplantation. Similarly, apart from ethical issues associated with disruption of inner cell mass, ESCs and ELSCs are also sources of HSCs as a therapeutic option to be utilized in SC transplantation. The generation of HSCs from iPSCs through hematopoietic–endothelial transition will be therapeutic options during times of inadequate availability of compatible donors. On the other hand, non-HSCs and MSCs are possible to use as coinfusion to support engraftment of transplants, hematologic reconstitution, and manage GVHD posttransplantation. Auto-HSCT and allo-HSCT are the major cellular therapeutic options to treat leukemia. The lower relapse risk, blast crisis, TKI-intolerant patients in the CP and at higher risk of disease, and higher relapse risk are indications to utilize allo-HSCT rather than auto-HSCT to treat different types of leukemia. Likewise, primary refractory sensitivity to relapse and positive PET are basic indications to prefer auto-HSCT to allo-HSCT in treating both multiple myeloma and HL. Therefore, allo-HSCT is a more applicable standard cellular therapeutic option than auto-HSCT for many classes of leukemia.

Abbreviations

Allo-HSCT, allogeneic hematopoietic stem–cell transplantation; auto-HSCT, autologous HSCT; CML, chronic myeloid leukemia; GVHD, graft-versus-host disease; ESCs, embryonic SCs; iPSCs, induced pluripotent SCs;MSCs, mesenchymal SCs; PBSCs, peripheral blood SCsVSELSCs, very small embryonic-like SCs.

Acknowledgments

The authors acknowledge Mrs Yonas Akalu for proofreading, language editing, and grammatical corrections to improve this review article.

Author Contributions

All authors made a significant contribution to the work reported, whether in conception, study design, execution, acquisition of data, analysis, interpretation, or all those areas; took part in drafting, revising, or critically reviewing the article; gave final approval to the version to be published; have agreed on the journal to which the article has been submitted; and agree to be accountable for all aspects of the work.

Disclosure

The authors declare that they have no competing interests.

References

1. Hawsawi YM, Al-Zahrani F, Mavromatis C, Baghdadi MA, Saggu S, Oyouni AAA. Stem cell applications for treatment of cancer and autoimmune diseases: its promises, obstacles, and future perspectives. Technol Cancer Res Treat. 2018;17:1533033818806910. doi:10.1177/1533033818806910

2. Weissman IL. Stem cells: units of development, units of regeneration, and units in evolution. cell. 2000;100(1):157–168. doi:10.1016/S0092-8674(00)81692-X

3. Pérez LM, de Lucas B, Gálvez BG. Unhealthy stem cells: when health conditions upset stem cell properties. Cell Physiol Biochem. 2018;46(5):1999–2016. doi:10.1159/000489440

4. Zakrzewski W, Dobrzyński M, Szymonowicz M, Rybak Z. Stem cells: past, present, and future. Stem Cell Res Ther. 2019;10(1):1–22. doi:10.1186/s13287-019-1165-5

5. He Q, Li J, Bettiol E, Jaconi ME. Embryonic stem cells: new possible therapy for degenerative diseases that affect elderly people. J Gerontol a Biol Sci Med Sci. 2003;58(3):M279–M87. doi:10.1093/gerona/58.3.M279

6. Tamura R, Toda M. Stem cell-based therapies for neurological disorders. AIMS Cell Tissue Eng. 2018;2(1):24. doi:10.3934/celltissue.2018.1.24

7. Inoue A, Kobayashi CI, Shinohara H, et al. Chronic myeloid leukemia stem cells and molecular target therapies for overcoming resistance and disease persistence. Int J Hematol. 2018;108(4):365–370. doi:10.1007/s12185-018-2519-y

8. Talleur AC, Flerlage JE, Shook DR, et al. Autologous hematopoietic cell transplantation for the treatment of relapsed/refractory pediatric, adolescent, and young adult Hodgkin lymphoma: a single institutional experience. Bone Marrow Transplant. 2020;1–10.

9. Zain J. Questions for stem cell transplant in peripheral T-cell non-Hodgkin’s lymphomas. Clin Investig (Lond). 2014;4(2):131–146. doi:10.4155/cli.13.133

10. Morgan RA, Gray D, Lomova A, Kohn DB. Hematopoietic stem cell gene therapy: progress and lessons learned. Cell Stem Cell. 2017;21(5):574–590. doi:10.1016/j.stem.2017.10.010

11. Gyurkocza B, Rezvani A, Storb RF. Allogeneic hematopoietic cell transplantation: the state of the art. Expert Rev Hematol. 2010;3(3):285–299. doi:10.1586/ehm.10.21

12. Halme DG, Kessler DA. FDA regulation of stem-cell–based therapies. Mass Med Soc. 2006;2:254.

13. Nirmalanandhan VS, Sittampalam GS. Stem cells in drug discovery, tissue engineering, and regenerative medicine: emerging opportunities and challenges. J Biomol Screen. 2009;14(7):755–768. doi:10.1177/1087057109336591

14. Biehl JK, Russell B. Introduction to stem cell therapy. J Cardiovasc Nurs. 2009;24(2):98. doi:10.1097/JCN.0b013e318197a6a5

15. Hima Bindu A, Srilatha B. Potency of various types of stem cells and their transplantation. J Stem Cell Res Ther. 2011;1:115.

16. Mimeault M, Hauke R, Batra SK. Stem cells: a revolution in therapeutics—recent advances in stem cell biology and their therapeutic applications in regenerative medicine and cancer therapies. Clin Pharmacol Ther. 2007;82(3):252–264. doi:10.1038/sj.clpt.6100301

17. Kumar R. Stem cell transplantation: indian perspective. J Ind Acad Clinic Med. 2002;3:182–188.

18. Corradini P, Cavo M, Lokhorst H, et al. Molecular remission after myeloablative allogeneic stem cell transplantation predicts a better relapse-free survival in patients with multiple myeloma. Blood. 2003;102(5):1927–1929. doi:10.1182/blood-2003-01-0189

19. Holtick U, Albrecht M, Chemnitz JM, et al. Comparison of bone marrow versus peripheral blood allogeneic hematopoietic stem cell transplantation for hematological malignancies in adults—a systematic review and meta-analysis. Crit Rev Oncol Hematol. 2015;94(2):179–188. doi:10.1016/j.critrevonc.2014.12.007

20. Kriegsmann K, Wuchter P. Mobilization and collection of peripheral blood stem cells in adults: focus on timing and benchmarking. Stem Cell Mobilization. 2019;1:41–58.

21. Fitch SR, Kapeni C, Tsitsopoulou A, et al. Gata3 targets Runx1 in the embryonic haematopoietic stem cell niche. IUBMB Life. 2019;3:24.

22. Kuruca SE, Çelik DD, Özerkan D, Erdemir G. Characterization and Isolation of Very Small Embryonic-like (VSEL) Stem Cells Obtained from Various Human Hematopoietic Cell Sources. Stem Cell Rev Rep. 2019;1–13.

23. Bhartiya D, Patel H, Ganguly R, et al. Novel insights into adult and cancer stem cell biology. Stem Cells Dev. 2018;27(22):1527–1539. doi:10.1089/scd.2018.0118

24. Petinati N, Drize N, Sats N, et al. Recovery of donor hematopoiesis after graft failure and second hematopoietic stem cell transplantation with intraosseous administration of mesenchymal stromal cells. Stem Cells Int. 2018;2018.

25. Tobin LM, Healy M, English K, Mahon BP. Human mesenchymal stem cells suppress donor CD4+ T cell proliferation and reduce pathology in a humanized mouse model of acute graft‐versus‐host disease. Clin Exp Immunol. 2013;172(2):333–348. doi:10.1111/cei.12056

26. Crippa S, Santi L, Bosotti R, Porro G, Bernardo ME. Bone marrow-derived mesenchymal stromal cells: a novel target to optimize hematopoietic stem cell transplantation protocols in hematological malignancies and rare genetic disorders. J Clin Med. 2020;9(1):2. doi:10.3390/jcm9010002

27. Cibelli JB, Kiessling AA, Cunniff K, Richards C, Lanza RP, West MD. Rapid communication: somatic cell nuclear transfer in humans: pronuclear and early embryonic development. e-Biomed. 2001;2(5):25–31. doi:10.1089/152489001753262168

28. Mehat MS, Sundaram V, Ripamonti C, et al. Transplantation of human embryonic stem cell-derived retinal pigment epithelial cells in macular degeneration. Ophthalmology. 2018;125(11):1765–1775. doi:10.1016/j.ophtha.2018.04.037

29. Prentice DA. Adult Stem Cells: successful Standard for Regenerative Medicine. Circ Res. 2019;124(6):837–839. doi:10.1161/CIRCRESAHA.118.313664

30. Bhartiya D. The need to revisit the definition of mesenchymal and adult stem cells based on their functional attributes. Stem Cell Res Ther. 2018;9(1):78. doi:10.1186/s13287-018-0833-1

31. Wang M, Wang H, Wen Y, et al. MEIS2 regulates endothelial to hematopoietic transition of human embryonic stem cells by targeting TAL1. Stem Cell Res Ther. 2018;9(1):340. doi:10.1186/s13287-018-1074-z

32. Lee JY, Kim M, Heo H-R, et al. Inhibition of microRNA-221 and 222 enhances hematopoietic differentiation from human pluripotent stem cells via c-kit upregulation. Mol Cells. 2018;41(11):971.

33. Wang F, Zhao W, Kong N, Cui W, Chai L. The next new target in leukemia: the embryonic stem cell gene SALL4. Mol Cell Oncol. 2014;1(4):e969169. doi:10.4161/23723548.2014.969169

34. Liu SP, Fu RH, Huang YC, et al. Induced pluripotent stem (iPS) cell research overview. Cell Transplant. 2011;20(1):15–19. doi:10.3727/096368910X532828

35. Ferreira AF, Calin GA, Picanço-Castro V, Kashima S, Covas DT, de Castro FA. Hematopoietic stem cells from induced pluripotent stem cells–considering the role of microRNA as a cell differentiation regulator. J Cell Sci. 2018;131(4):jcs203018.

36. Preynat-Seauve O, Krause K-H, editors. Stem cell sources for regenerative medicine: the immunological point of view. In: Seminars in Immunopathology. Springer; 2011.

37. Roubelakis MG. Introduction to Stem Cell Principles and Biology. In: Digestive System Diseases. Springer; 2019:7–20.

38. Seki T, Yuasa S, Oda M, et al. Generation of induced pluripotent stem cells from human terminally differentiated circulating T cells. Cell Stem Cell. 2010;7(1):11–14. doi:10.1016/j.stem.2010.06.003

39. Dimos JT, Rodolfa KT, Niakan KK, et al. Induced pluripotent stem cells generated from patients with ALS can be differentiated into motor neurons. science. 2008;321(5893):1218–1221. doi:10.1126/science.1158799

40. Loh Y-H, Agarwal S, Park I-H, et al. Generation of induced pluripotent stem cells from human blood. Blood. 2009;113(22):5476–5479. doi:10.1182/blood-2009-02-204800

41. Chao MP, Majeti R. Induced pluripotent stem cell modeling of malignant hematopoiesis. Exp Hematol. 2019;71:68–76. doi:10.1016/j.exphem.2019.01.002

42. Lange L, Hoffmann D, Schwarzer A, et al. Inducible forward programming of human pluripotent stem cells to hemato-endothelial progenitor cells with hematopoietic progenitor potential. Stem Cell Rep. 2019.

43. Masouridi-Levrat S, Simonetta F, Chalandon Y. Immunological basis of bone marrow failure after allogeneic hematopoietic stem cell transplantation. Front Immunol. 2016;7:362. doi:10.3389/fimmu.2016.00362

44. Ferreira AF, Calin GA, Picanço-Castro V, Kashima S, Covas DT, de Castro FA. Hematopoietic stem cells from induced pluripotent stem cells–considering the role of microRNA as a cell differentiation regulator. J Cell Sci. 2018;131(4):4. doi:10.1242/jcs.203018

45. Kaushik A, Bhartiya D. Pluripotent very small embryonic-like stem cells in adult testes–an alternate premise to explain testicular germ cell tumors. Stem Cell Rev Rep. 2018;14(6):793–800. doi:10.1007/s12015-018-9848-3

46. Lahlil R, Scrofani M, Barbet R, Tancredi C, Aries A, Hénon P. VSELs maintain their pluripotency and competence to differentiate after enhanced ex vivo expansion. Stem Cell Rev Rep. 2018;14(4):510–524. doi:10.1007/s12015-018-9821-1

47. Ratajczak MZ, Ratajczak J, Kucia M. Very small embryonic-like stem cells (VSELs) An Update and Future Directions. Circ Res. 2019;124(2):208–210. doi:10.1161/CIRCRESAHA.118.314287

48. !!! INVALID CITATION !!!

49. Eljaszewicz A, Bolkun L, Grubczak K, et al. Very small embryonic-like stem cells, endothelial progenitor cells, and different monocyte subsets are effectively mobilized in acute lymphoblastic leukemia patients after G-CSF Treatment. Stem Cells Int. 2018;2018:256.

50. Sasaki M, Abe R, Fujita Y, Ando S, Inokuma D, Shimizu H. Mesenchymal stem cells are recruited into wounded skin and contribute to wound repair by transdifferentiation into multiple skin cell type. J Immunol. 2008;180(4):2581–2587. doi:10.4049/jimmunol.180.4.2581

51. Zhu Y, Sun Z, Han Q, et al. Human mesenchymal stem cells inhibit cancer cell proliferation by secreting DKK-1. Leukemia. 2009;23(5):925. doi:10.1038/leu.2008.384

52. Nakano M, Nagaishi K, Konari N, et al. Bone marrow-derived mesenchymal stem cells improve diabetes-induced cognitive impairment by exosome transfer into damaged neurons and astrocytes. Sci Rep. 2016;6(1):24805. doi:10.1038/srep24805

53. Hartley CP, Carrillo-Polanco LF, Rowan DJ, Hagen CE. Colonic graft-vs.-host disease in autologous versus allogeneic transplant patients: earlier onset, more apoptosis, and lack of regulatory T-cell attenuation. Modern Pathol. 2018;31(10):1619–1626. doi:10.1038/s41379-018-0078-7

54. Miura Y. Human bone marrow mesenchymal stromal/stem cells: current clinical applications and potential for hematology. Int J Hematol. 2016;103(2):122–128. doi:10.1007/s12185-015-1920-z

55. de la Guardia RD, Lopez-Millan B, Roca-Ho H, et al. Bone marrow mesenchymal stem/stromal cells from risk-stratified acute myeloid leukemia patients are anti-inflammatory in in vivo preclinical models of hematopoietic reconstitution and severe colitis. haematologica. 2018;196568.

56. Saudemont A, Madrigal JA. Immunotherapy after hematopoietic stem cell transplantation using umbilical cord blood-derived products. Cancer Immunol Immunother. 2017;66(2):215–221. doi:10.1007/s00262-016-1852-3

57. Joshi SS, Tarantolo SR, Kuszynski CA, Kessinger A. Antitumor therapeutic potential of activated human umbilical cord blood cells against leukemia and breast cancer. Clin Cancer Res. 2000;6(11):4351–4358.

58. Yabe T, Azuma F, Kashiwase K, et al. HLA-DPB1 mismatch induces a graft-versus-leukemia effect without severe acute GVHD after single-unit umbilical cord blood transplantation. Leukemia. 2018;32(1):168–175. doi:10.1038/leu.2017.202

59. Brunstein CG, Cutler CS, DeFor TE, et al. Matching at HLA-C improved the outcomes after double umbilical cord blood transplantation for recipients of 2–4/6 HLA-matched grafts. Biol Blood Marrow Transplant. 2017;23(1):126. doi:10.1016/j.bbmt.2016.10.018

60. Baron F, Labopin M, Ruggeri A, et al. Unrelated cord blood transplantation for adult patients with acute myeloid leukemia: higher incidence of acute graft-versus-host disease and lower survival in male patients transplanted with female unrelated cord blood—a report from Eurocord, the acute leukemia working party, and the cord blood committee of the cellular therapy and immunobiology working party of the european group for blood and marrow transplantation. J Hematol Oncol. 2015;8(1):107.

61. Ballen K. Umbilical cord blood transplantation: challenges and future directions. Stem Cells Transl Med. 2017;6(5):1312–1315. doi:10.1002/sctm.17-0069

62. Bhatt VR. Allogeneic stem cell transplantation for non-Hodgkin lymphoma. Curr Hematol Malig Rep. 2016;11(3):196–207. doi:10.1007/s11899-016-0319-0

63. Gagelmann N, Bacigalupo A, Rambaldi A, et al. Haploidentical stem cell transplantation with posttransplant cyclophosphamide therapy vs other donor transplantations in adults with hematologic cancers: a systematic review and meta-analysis. JAMA Oncol. 2019;5(12):1739–1748. doi:10.1001/jamaoncol.2019.3541

64. Baumeister SH, Rambaldi B, Shapiro RM, Romee R. Key aspects of the immunobiology of haploidentical hematopoietic cell transplantation. Front Immunol. 2020;11:191. doi:10.3389/fimmu.2020.00191

65. Bondanza A, Ruggeri L, Noviello M, et al. Beneficial role of CD8+ T-cell reconstitution after HLA-haploidentical stem cell transplantation for high-risk acute leukaemias: results from a clinico-biological EBMT registry study mostly in the T-cell-depleted setting. Bone Marrow Transplant. 2019;54(6):867–876. doi:10.1038/s41409-018-0351-x

66. Sahasrabudhe K, Otto M, Hematti P, Kenkre V. TCR αβ+/CD19+ cell depletion in haploidentical hematopoietic allogeneic stem cell transplantation: a review of current data. Leuk Lymphoma. 2019;60(3):598–609. doi:10.1080/10428194.2018.1485905

67. Merli P, Algeri M, Galaverna F, et al. Immune Modulation Properties of Zoledronic Acid on TcRγδ T-Lymphocytes After TcRαβ/CD19-Depleted Haploidentical Stem Cell Transplantation: an analysis on 46 Pediatric Patients Affected by Acute Leukemia. Front Immunol. 2020;11:699. doi:10.3389/fimmu.2020.00699

68. Raiola AM, Risitano A, Sacchi N, et al. Impact of HLA disparity in haploidentical bone marrow transplantation followed by high-dose cyclophosphamide. Biol Blood Marrow Transplant. 2018;24(1):119–126. doi:10.1016/j.bbmt.2017.10.002

69. Lydon H, Brooks R, McCaskie A, Henson F. Peripheral mononuclear blood cell apheresis in a preclinical ovine model. BMC Vet Res. 2018;14(1):47. doi:10.1186/s12917-018-1332-4

70. Broughton KM, Sussman MA. Enhancement strategies for cardiac regenerative cell therapy: focus on adult stem cells. Circ Res. 2018;123(2):177–187. doi:10.1161/CIRCRESAHA.118.311207

71. Or R, Shapira MY, Resnick I, et al. Nonmyeloablative allogeneic stem cell transplantation for the treatment of chronic myeloid leukemia in first chronic phase. Blood. 2003;101(2):441–445. doi:10.1182/blood-2002-02-0535

72. Slavin S, Nagler A, Naparstek E, et al. Nonmyeloablative stem cell transplantation and cell therapy as an alternative to conventional bone marrow transplantation with lethal cytoreduction for the treatment of malignant and nonmalignant hematologic diseases. Blood. 1998;91(3):756–763. doi:10.1182/blood.V91.3.756

73. Chow A, Lucas D, Hidalgo A, et al. Bone marrow CD169+ macrophages promote the retention of hematopoietic stem and progenitor cells in the mesenchymal stem cell niche. J Exp Med. 2011;208(2):261–271. doi:10.1084/jem.20101688

74. Vellenga E, van Putten W, Ossenkoppele GJ, et al. Autologous peripheral blood stem cell transplantation for acute myeloid leukemia. Blood. 2011;118(23):6037–6042. doi:10.1182/blood-2011-07-370247

75. Javarappa KK, Tsallos D, Heckman CA. A Multiplexed Screening Assay to Evaluate Chemotherapy-Induced Myelosuppression Using Healthy Peripheral Blood and Bone Marrow. SLAS DISCOVER. 2018;23(7):687–696.

76. Kansagra A, Gonsalves WI, Gertz MA, et al. Analysis of clinical factors and outcomes associated with nonuse of collected peripheral blood stem cells for autologous stem cell transplants in transplant-eligible patients with multiple myeloma. Biol Blood Marrow Transplant. 2018;24(10):2127–2132. doi:10.1016/j.bbmt.2018.04.007

77. Ruggeri A, Labopin M, Bacigalupo A, et al. Bone marrow versus mobilized peripheral blood stem cells in haploidentical transplants using posttransplantation cyclophosphamide. Cancer. 2018;124(7):1428–1437. doi:10.1002/cncr.31228

78. Orbach D, Hojjat-Assari S, Doz F, et al. Peripheral blood stem cell collection in 24 low-weight infants: experience of a single centre. Bone Marrow Transplant. 2003;31(3):171. doi:10.1038/sj.bmt.1703825

79. Passegué E, Wagers AJ, Giuriato S, Anderson WC, Weissman IL. Global analysis of proliferation and cell cycle gene expression in the regulation of hematopoietic stem and progenitor cell fates. J Exp Med. 2005;202(11):1599–1611. doi:10.1084/jem.20050967

80. Rahmat LT, Nguyen A, Abdulhaq H, Prakash S, Logan AC, Mannis GN. Venetoclax in combination with decitabine for relapsed T-Cell acute lymphoblastic leukemia after allogeneic hematopoietic cell transplant. Case Rep Hematol. 2018;2018.

81. Roche-Lestienne C, Soenen-Cornu V, Grardel-Duflos N, et al. Several types of mutations of the Abl gene can be found in chronic myeloid leukemia patients resistant to STI571, and they can pre-exist to the onset of treatment. Blood. 2002;100(3):1014–1018. doi:10.1182/blood.V100.3.1014

82. Miyauchi M, Koya J, Arai S, et al. ADAM8 is an antigen of tyrosine kinase inhibitor-resistant chronic myeloid leukemia cells identified by patient-derived induced pluripotent stem cells. Stem Cell Rep. 2018;10(3):1115–1130. doi:10.1016/j.stemcr.2018.01.015

83. Mens MM, Ghanbari M. Cell cycle regulation of stem cells by microRNAs. Stem Cell Rev Rep. 2018;14(3):309–322. doi:10.1007/s12015-018-9808-y

84. Sell S. On the stem cell origin of cancer. Am J Pathol. 2010;176(6):2584–2594. doi:10.2353/ajpath.2010.091064

85. Harada K, Doki N, Aoki J, et al. Outcomes after allogeneic hematopoietic stem cell transplantation in patients with acute myeloid leukemia harboring t (7; 11)(p15; p15). haematologica. 2018;103(2):e69. doi:10.3324/haematol.2017.179804

86. McSweeney PA, Niederwieser D, Shizuru JA, et al. Hematopoietic cell transplantation in older patients with hematologic malignancies: replacing high-dose cytotoxic therapy with graft-versus-tumor effects. Blood. 2001;97(11):3390–3400. doi:10.1182/blood.V97.11.3390

87. Morrison CF, Pai AL, Martsolf D. Facilitators and barriers to self-management for adolescents and young adults following a hematopoietic stem cell transplant. J Pediatr Oncol Nurs. 2018;35(1):36–42. doi:10.1177/1043454217723864

88. Thomas ED, Blume KG. Historical markers in the development of allogeneic hematopoietic cell transplantation. Biol Blood Marrow Transplant. 1999;5(6):341–346. doi:10.1016/S1083-8791(99)70010-8

89. Khouri IF, Curbelo IF, Turturro F, et al. Ipilimumab plus lenalidomide after allogeneic and autologous stem cell transplantation for patients with lymphoid malignancies. Clin Cancer Res. 2018;24(5):1011–1018. doi:10.1158/1078-0432.CCR-17-2777

90. Dos Santos DMC, Saliba RM, Patel R, et al. Age is a prognostic factor for the overall survival of multiple myeloma patients undergoing upfront autologous hematopoietic stem cell transplantation. Biol Blood Marrow Transplant. 2019;2:15–25.

91. Hammami MB, Talkin R, Al-Taee AM, Schoen MW, Goyal SD, Lai J-P. Autologous graft-versus-host disease of the gastrointestinal tract in patients with multiple myeloma and hematopoietic stem cell transplantation. Gastroenterol Res. 2018;11(1):52. doi:10.14740/gr925w

92. Krishnan A, Molina A, Zaia J, et al. Durable remissions with autologous stem cell transplantation for high-risk HIV-associated lymphomas. Blood. 2005;105(2):874–878. doi:10.1182/blood-2004-04-1532

93. Shimoni A, Hardan I, Shem-Tov N, et al. Allogeneic hematopoietic stem-cell transplantation in AML and MDS using myeloablative versus reduced-intensity conditioning: the role of dose intensity. Leukemia. 2006;20(2):322. doi:10.1038/sj.leu.2404037

94. Kröger N, Schwerdtfeger R, Kiehl M, et al. Autologous stem cell transplantation followed by a dose-reduced allograft induces high complete remission rate in multiple myeloma. Blood. 2002;100(3):755–760. doi:10.1182/blood-2002-01-0131

95. Martino R, Caballero M, Simón JAP, et al. Evidence for a graft-versus-leukemia effect after allogeneic peripheral blood stem cell transplantation with reduced-intensity conditioning in acute myelogenous leukemia and myelodysplastic syndromes. Blood. 2002;100(6):2243–2245. doi:10.1182/blood-2002-02-0400

96. Giralt S, Estey E, Albitar M, et al. Engraftment of allogeneic hematopoietic progenitor cells with purine analog-containing chemotherapy: harnessing graft-versus-leukemia without myeloablative therapy. Blood. 1997;89(12):4531–4536. doi:10.1182/blood.V89.12.4531

97. Soverini S, Mancini M, Bavaro L, Cavo M, Martinelli G. Chronic myeloid leukemia: the paradigm of targeting oncogenic tyrosine kinase signaling and counteracting resistance for successful cancer therapy. Mol Cancer. 2018;17(1):1–15. doi:10.1186/s12943-018-0780-6

98. Jabbour E, Kantarjian H. Chronic myeloid leukemia: 2018 update on diagnosis, therapy and monitoring. Am J Hematol. 2018;93(3):442–459. doi:10.1002/ajh.25011

99. Jabbour E, Kantarjian H. Chronic myeloid leukemia: 2020 update on diagnosis, therapy and monitoring. Am J Hematol. 2020;95(6):691–709. doi:10.1002/ajh.25792

100. Copland M. Is There a Role for Dose Modification of TKI Therapy in CML? Curr Hematol Malig Rep. 2019;14(4):337–345. doi:10.1007/s11899-019-00524-w

101. Tan FH, Putoczki TL, Stylli SS, Luwor RB. Ponatinib: a novel multi-tyrosine kinase inhibitor against human malignancies. Onco Targets Ther. 2019;12:635. doi:10.2147/OTT.S189391

102. Bhamidipati PK, Kantarjian H, Cortes J, Cornelison AM, Jabbour E. Management of imatinib-resistant patients with chronic myeloid leukemia. Ther Adv Hematol. 2013;4(2):103–117. doi:10.1177/2040620712468289

103. Cerveira N, Ferreira RB, Bizarro S, et al. Ponatinib induces a sustained deep molecular response in a chronic myeloid leukaemia patient with an early relapse with a T315I mutation following allogeneic hematopoietic stem cell transplantation: a case report. BMC Cancer. 2018;18(1):1–5. doi:10.1186/s12885-018-5100-4

104. Chen X, Cao Z, Hu J. Advances in allogeneic hematopoietic stem cell transplantation for chronic myelogenous leukemia–review. Zhongguo Shi Yan Xue Ye Xue Za Zhi. 2019;27(4):1334–1338. doi:10.19746/j.cnki.issn.1009-2137.2019.04.057

105. Barba P, Martino R, Zhou Q, et al. CD34+ cell selection versus reduced-intensity conditioning and unmodified grafts for allogeneic hematopoietic cell transplantation in patients age> 50 years with acute myelogenous leukemia and myelodysplastic syndrome. Biol Blood Marrow Transplant. 2018;24(5):964–972. doi:10.1016/j.bbmt.2017.12.804

106. Balduzzi A, Dalle J-H, Wachowiak J, et al. Transplantation in children and adolescents with acute lymphoblastic leukemia from a matched donor versus an HLA-identical sibling: is the outcome comparable? Results from the international bfm all SCT 2007 study. Biol Blood Marrow Transplant. 2019;25(11):2197–2210. doi:10.1016/j.bbmt.2019.07.011

107. Santoro N, Ruggeri A, Labopin M, et al. Unmanipulated haploidentical stem cell transplantation in adults with acute lymphoblastic leukemia: a study on behalf of the Acute Leukemia Working Party of the EBMT. J Hematol Oncol. 2017;10(1):113. doi:10.1186/s13045-017-0480-5

108. Chen C-T, Gau J-P, Liu J-H, Chiou T-J, Hsiao L-T, Liu Y-C. Early achievement of full donor chimerism after allogeneic hematopoietic stem cell transplantation predicts lower relapse risk in patients with acute lymphoblastic leukemia. J Chin Med Assoc. 2018;81(12):1038–1043. doi:10.1016/j.jcma.2018.06.005

109. Dreger P, Corradini P, Kimby E, et al. Indications for allogeneic stem cell transplantation in chronic lymphocytic leukemia: the EBMT transplant consensus. Leukemia. 2007;21(1):12. doi:10.1038/sj.leu.2404441

110. Wang T, Gao L, Hu X, et al. Chimeric antigen receptor-modified donor lymphocyte infusion improves the survival of acute lymphoblastic leukemia patients with relapsed diseases after allogeneic hematopoietic stem cell transplantation. J Immunother. 2019;42(3):81–88. doi:10.1097/CJI.0000000000000257

Creative Commons License © 2020 The Author(s). This work is published and licensed by Dove Medical Press Limited. The full terms of this license are available at https://www.dovepress.com/terms.php and incorporate the Creative Commons Attribution - Non Commercial (unported, v3.0) License. By accessing the work you hereby accept the Terms. Non-commercial uses of the work are permitted without any further permission from Dove Medical Press Limited, provided the work is properly attributed. For permission for commercial use of this work, please see paragraphs 4.2 and 5 of our Terms.