Hematopoietic stem cell transplantation and cellular therapy


Hematopoietic stem cell transplantation (HSCT) is an accepted treatment of choice for a variety of malignant and nonmalignant disorders. Tables 30.1 and 30.2 list indications for allogeneic and autologous HSCT. Most allogeneic transplants perform.ed in patients <20 years old are for acute leukemias (43%) or nonmalignant indications (35%). Preparation, or “conditioning,” for HSCT involves the delivery of high-dose chemotherapy (HDC) with or without radiation to ablate or reduce hematopoiesis and to provide sufficient immunosuppression to allow donor cell engraftment.

Table 30.1
Indications for allogeneic hematopoietic stem cell transplantation in children.
Malignant disorders
This group includes patients under 21 years of age with any of the following:
AML

  • 1.

    High-risk CR1 a , defined as:

    • a.

      Having preceding MDS or

    • b.

      High-risk cytogenetics: del (5q) -5, -7, abn (3q), t(6;9) complex karyotype (≥5 abnormalities) or

    • c.

      Requiring >1 cycle of chemotherapy to obtain CR (this includes any clinical or radiographic evidence of progressive extramedullary AML)

    • d.

      FAB classification: M6

    • e.

      The presence of high (>0.4) allelic ratio FLT3-ITD

    • f.

      Patients with therapy-related AML

  • 2.

    Second or greater CR

  • 3.

    Responsive disease that is not in remission (relapse risk is very high)

ALL

  • 1.

    High-risk CR1 b , defined as:

    • a.

      infants with MLL rearrangements (KMT2A-r), <6 month of age with high-risk characteristics (>300,000 white blood cells/μL); or

    • b.

      severe hypodiploidy (<44 chromosomes and/or a DNA index of <0.81); or

    • c.

      M3 bone marrow at end of induction; or

    • d.

      persistent MRD≥0.01% after consolidation.

  • 2.

    High-risk CR2, defined as:

    • a.

      Ph+ALL; or

    • b.

      bone marrow relapse <36 months from induction; or

    • c.

      T-lineage relapse at any time; or

    • d.

      early isolated CNS relapse (<18 months from diagnosis); or

    • e.

      late relapse (>36 m) with ≥0.1% MRD after induction.

  • 3.

    Any third or subsequent CR

Ph+CML

  • 1.

    chronic unstable phase c ;

  • 2.

    early accelerated phase; or

  • 3.

    chronic phase after treatment for blast crisis.

JMML
Lymphomas (second/subsequent complete remission, or partial remission)

  • 1.

    Hodgkin

  • 2.

    Non-Hodgkin

Myelodysplasia
Myelofibrosis
Nonmalignant disorders
Congenital

  • 1.

    Immunodeficiency and immune dysregulatory diseases:

    • a.

      SCIDs

    • b.

      WAS d

    • c.

      Chronic granulomatous disease

    • d.

      Familial HLH

    • e.

      X-linked hyper IgM (CD40 ligand deficiency)

    • f.

      IPEX syndrome

  • 2.

    Hematologic disorders:

    • a.

      Hemoglobinopathies: sickle cell disease e and thalassemia

    • b.

      Fanconi anemia f (with the evidence of marrow failure or myelodysplasia)

    • c.

      Shwachman–Diamond syndrome f (with the evidence of marrow failure or myelodysplasia)

    • d.

      Kostmann agranulocytosis (if failing G-CSF or progressed to MDS) g

    • e.

      Diamond–Blackfan anemia nonresponsive to steroids h

    • f.

      Dyskeratosis congenita with marrow failure f

    • g.

      CAMT

    • h.

      Chediak–Higashi syndrome

    • i.

      LAD I: severe phenotype defined by the absence (≤1%) of CD11b/CD18 expression on neutrophils

  • 3.

    Lysosomal storage diseases i :

    • a.

      X-ALD with all of the following:

      • i.

        Evidence of brain demyelination (i.e., measurable MRI severity score ≥1) and/or gadolinium enhancement by MRI brain

      • ii.

        Brain demyelination MRI severity score <9

    • b.

      MLD with all of the following:

      • i.

        Confirmed with both leukocyte arylsulfatase A deficiency and increased urinary sulfatides to exclude pseudo-deficiency

      • ii.

        Presymptomatic juvenile or adult form of the disease

    • c.

      Infantile GLD (Krabbe disease) must be asymptomatic

    • d.

      Gaucher disease (Type III): with new or deteriorating neurological and/or pulmonary symptoms while on enzyme replacement therapy

    • e.

      Hurler syndrome

  • 4.

    Osteopetrosis: malignant infantile osteopetrosis or osteopetrosis with CA II deficiency

Acquired

  • 1.

    Severe aplastic anemia

  • 2.

    Paroxysmal nocturnal hemoglobinuria (failing complement blockade therapy)

  • 3.

    Acquired HLH relapse after initial therapy

Abbreviations: ALL , Acute lymphocytic leukemia; AML , acute myelogenous leukemia; CAR , chimeric antigen receptor; CA , carbonic anhydrase; COG , Children Oncology Group; CML , chronic myelogenous leukemia; CAMT , congenital amegakaryocytic thrombocytopenia; ETP , early T-cell precursor; CR1 , first complete remission; G-CSF , granulocyte colony-stimulating factor; HLH , hemophagocytic lymphohistiocytosis; HLA , human leukocyte antigen; HSCT , hematopoietic stem cell transplantation; IPEX syndrome , immune dysregulation, polyendocrinopathy, enteropathy, X-linked syndrome; JMML , juvenile myelo-monocytic leukemia; LAD I , leukocyte adhesion deficiency type I; MRA , magnetic resonance angiography; MRI , magnetic resonance imaging; MRD , minimal residual disease; MLD , metachromatic leukodystrophy; MLL , myeloid/lymphoid or mixed lineage leukemia; MDS , myelodysplasia; NGS , next-generation sequencing; OS , overall survival; CR2 , second complete remission; SCIDs , severe combined immunodeficiency syndromes; SCD , sickle cell disease; TKIs , tyrosine kinase inhibitors; WAS , Wiskott–Aldrich syndrome.

a The high-risk group as defined represents approximately 30% of patients and has a predicted OS<35%. This group additionally, may include non-low-risk cytogenetics with positive (≥0.1%) MRD at the end of Induction I [low-risk cytogenetics is defined as inc(16)/t(16;16) or (8;21) cytogenetics or NPM or CEBPα mutations]. This group of patients should optimally receive HSCT, if possible, after Intensification I. The COG is currently reexamining the risk stratification for newly diagnosed AML (AAML1831 study). Indications for HSCT in CR1 are to be based on high-risk cytogenetics and molecular markers along with response based on MRD (after Induction II). Any patient without a good risk marker who has MRD will proceed to HSCT. Patients without a high-risk marker who has no MRD will not proceed to HSCT. Under this study the following groups are classified as high-risk patients and will receive HSCT in CR1 (if a donor available):

  • FLT3/ITD+ with allelic ratio >0.1 without bZIP CEBPA, NPM1;

  • FLT3/ITD+ with allelic ratio >0.1 with concurrent bZIP CEBPA or NPM1 and with evidence of residual AML (MRD≥0.05%) at end of Induction I;

  • the presence of RAM phenotype or unfavorable prognostic markers (other than FLT3/ITD) per cytogenetics, FISH, NGS results, regardless of favorable genetic markers, MRD status or FLT3/ITD mutation;

  • AML without favorable or unfavorable cytogenetic or molecular features, but with evidence of residual AML (MRD≥0.05%) at the end of Induction I; and

  • the presence of a non-ITD FLT3 activating mutation and positive MRD (≥0.05%) at the end of Induction I regardless of the presence of favorable genetic markers

b Investigators differ regarding the optimum treatment and may consider HSCT for ALL in CR1 for Ph+ ALL with available HLA-matched sibling; MLL (KMT2A) rearrangement with slow early response [defined as having M2 (5–25% blasts) or M3 (>25% blasts on bone marrow examination on Day 14 of induction therapy)]; intrachromosomal amplification of chromosome 21(iAMP21); ETP-ALL, ALL with positive >or =0.01% MRD especially at the end of consolidation. The approach to therapy of B-ALL is rapidly evolving with the recent availability of CAR-T cell therapy and is likely to modify the indications and utility of HSCT for treatment of B-ALL.

c Unstable phase as defined with longitudinal Q RT-PCR for BCR-ABL in response to TKIs treatment; unlike adults, in children, investigators differ regarding the optimum treatment and may consider HSCT, if optimal, donor is available, regardless of the response to TKIs treatment. However, when to transplant children with CML remains controversial.

d Indications for HSCT using unrelated donor (or cord blood) in WAS include; refractory or symptomatic transfusion dependent thrombocytopenia, severe autoimmune manifestations not controlled on medical therapy, life-threatening or recurrent serious infections in the past requiring hospital admissions while on standard prophylaxis.

e Indications for HSCT in SCD may include history of one or more of the following: Recurrent vaso-occlusive crisis requiring hospitalizations or emergency room visits and narcotic use to control pain; evidence of SCD ischemia or pathology by cerebral MRI or cerebral MRA scan; history of elevated trans-cranial flow Doppler studies; recurrent acute chest syndrome, sickle nephropathy; Grade ≥1 avascular necrosis of 1≥ joint(s); red cell alloimmunization during transfusion therapy interfering with the ability to use transfusion therapy.

f Reduced intensity conditioning used due to poor tolerance to chemotherapy.

g G-CSF may be successful in treatment and avoiding HSCT. Requiring G-CSF therapy ≥10 µg/kg per day or marrow failure resulting in pancytopenia requiring transfusions is clear indication to consider HSCT from any suitable unrelated donor or cord blood.

h Transfusion dependance or failure of steroids are clear indications to consider HSCT from any suitable unrelated donor or cord blood.

i HSCT provides a population of cells with the capacity to produce the missing enzyme. Early transplantation is the goal so that enzyme replacement may occur before extensive central nervous system injury becomes evident.

Table 30.2
Indications for autologous hematopoietic stem cell transplantation.
Hematologic malignancies
  • 1.

    Non-Hodgkins lymphoma a

  • 2.

    Hodgkins disease a

  • 3.

    Acute promyelocytic leukemia (≥CR2)

Solid tumors
  • 1.

    Neuroblastoma (high-risk)

  • 2.

    Ewing’s sarcoma and PNET b

  • 3.

    Germ cell tumor a

  • 4.

    Brain tumors a,c

  • 5.

    Wilms tumor a

  • 6.

    Retinoblastoma a

Abbreviations: CR2 , Second complete remission; PNET , primitive neuroectodermal tumors.

a Relapsed or refractory disease but achieved at least partial response with conventional chemotherapy (i.e., chemosensitive disease).

b Metastatic disease to the lung only at presentation (practiced in Europe, controversial in North America).

c Used as an approach to avoid radiation in infants and young children.

The rationale for HDC is grounded in the finding of a steep dose–response curve for many chemotherapeutic agents. Most drugs exhibit a log–linear relationship between tumor cell kill and dose over a certain range, followed by flattening of the curve in the upper dose ranges. For this reason, small changes in dose can produce a significant change in response to chemotherapeutic agents. For many chemotherapeutic agents, the major dose-limiting toxicity is myelosuppression. Doses of chemotherapy sufficient to maximize killing of certain cancers cannot be delivered out of concern for permanent damage to the hematopoietic system. While hematopoietic growth factor support offers the potential to maximize the dose–response of standard-dose chemotherapy, allogeneic HSCT or autologous hematopoietic stem cell (HSC) rescue offers the opportunity to exceed marrow tolerance. This permits the delivery of a higher dose of chemotherapeutic agents, thus achieving a higher peak on the tumor-kill versus drug dose curve. For many agents a 3- to 10-fold increase in drug dose may result in a multiple log increase in tumor cell killing. In addition, for most high-dose conditioning regimens, multiple drugs are used in order to overcome tumor heterogeneity and drug resistance.

The most common type of HSCT to treat hematological malignancies and nonmalignant disorders is allogeneic, using a human leukocyte antigen (HLA)-matched histocompatible donor. Solid tumors have been treated with HDC followed by autologous HSC rescue with the concomitant use of hematopoietic growth factors such as granulocyte colony-stimulating factor (G-CSF) to reduce the duration of neutropenia caused by escalating doses of chemotherapy.

Table 30.3 lists the different sources of HSCs used for transplantation. Tables 30.4 and 30.5 list the advantages and disadvantages of allogeneic and autologous HSCT, respectively.

Table 30.3
Sources of hematopoietic stem cells for transplantation.
  • 1.

    Allogeneic bone marrow or PBSC using an HLA-matched sibling

  • 2.

    Allogeneic bone marrow or PBSC using matched [or partially matched (seven out of eight allele matched)] HLA family members if available

  • 3.

    Syngeneic bone marrow or PBSC (an identical twin)

  • 4.

    Allogeneic bone marrow or PBSC using HLA-matched or mismatched unrelated donors

  • 5.

    Autologous PBSCs: The number of circulating stem cells can be increased by the use of recombinant growth factors, which can increase stem cell numbers in peripheral blood prior to apheresis. This induces a more rapid hematopoietic recovery after transplantation

  • 6.

    Autologous bone marrow: only of autologous PBSC cannot be obtained

  • 7.

    UCB stem cells (always allogeneic). Using an HLA-matched sibling or an unrelated cord blood (single- or double units). Ex vivo expansion of cells to increase the number of CD34+ cells available for transplant has been employed experimentally

Abbreviations: HLA , Human leukocyte antigen; HSCT , hematopoietic stem cell transplantation; PBSC , peripheral blood stem cell; UCB , umbilical cord blood.

Table 30.4
Advantages and disadvantages of allogeneic hematopoietic stem cell transplantation.
Advantages Disadvantages
  • 1.

    Lower relapse rate

  • 2.

    Graft versus leukemia effect a

  • 1.

    Compatible donor availability limited (25% of siblings are compatible, minority groups less likely to have compatible unrelated donors)

  • 2.

    GVHD

  • 3.

    Increased risk of infections such as cytomegalovirus or adenovirus and interstitial pneumonia (this risk is 2.5 times greater in unrelated matched donors compared to matched siblings). Immune suppression due to GVHD prophylaxis or treatment drugs

  • 4.

    Risk of veno-occlusive disease (liver) or thrombotic microangiopathy (kidney) or other organ damage

Abbreviation: GVHD , Graft-versus-host disease.

a The relapse rate is 2.5 times lower in allogeneic recipients who have grade II–IV acute GVHD compared to recipients without GVHD. Leukemic cells have been reported to disappear during episodes of acute GVHD.

Table 30.5
Advantages and disadvantages of autologous hematopoietic stem cell transplantation.
Advantages Disadvantages
  • 1.

    No need for an allogeneic donor

  • 2.

    No GVHD

  • 3.

    Lower risk of opportunistic infection (e.g., CMV, P. jirovecii )

  • 1.

    Increased risk of relapse (approximately 50%)

  • 2.

    No graft versus leukemia effect

    • a.

      Rare risk of graft insufficiency posttransplant

Abbreviations: CMV , Cytomegalovirus; GVHD, graft-versus-host disease.

Allogeneic stem cell transplantation

Studies over the last several decades have shown that the degree of match between the donor and recipient major histocompatibility complex (MHC) genes has been directly related to critical outcomes such as rejection, infection, graft-versus-host disease (GVHD), and survival. Although newer approaches to mismatched and haploidentical HSCT have improved outcomes, all patients undergoing allogeneic HSCT require a detailed assessment of MHC genotypes.

Histocompatibility testing

The MHC genes are mapped within a region called HLA located on the short arm of chromosome 6. HLA antigens are responsible for the rejection of foreign objects from the body. There are at least six major HLA loci tested as needed to identify best donors for allogeneic HSCT—A, B, C, DR, DQ, and DP. These loci are divided into two groups: class I antigens, HLA-A, HLA-B, and HLA-C, and class II antigens, HLA-DR, HLA-DQ, and HLA-DP. They are segregated by haplotype from an individual father and mother. The class I molecules are composed of an α chain and β2 microglobulin, are highly polymorphic, and are expressed on most nucleated cells and on platelets. The HLA class II loci are involved in exogenous antigen processing. The HLA class II antigens are heterodimeric cell surface molecules formed by the α and β chains, each of which is polymorphic. HLA terminology is designated by the World Health Organization nomenclature committee for factors of the HLA system and is updated at regular intervals ( https://www.ebi.ac.uk/ipd/imgt/hla/ ).

To assess the degree of HLA compatibility, donor and recipient need to be HLA “typed.” Typing methods have included serological (antigen) or molecular (allele) (with low, intermediate, or high resolution) approaches. The broadest designation of HLA type is based on serologic typing, with the highest specificity based on the actual DNA sequence.

Serological tests are rarely used. The following techniques have been used for the identification of class I and II types:

  • Sequence-specific oligonucleotide probe

  • Sequence-specific primer

  • DNA sequencing [usually reserved to resolve any ambiguity that is not resolved by the previous methods. Routine next-generation sequencing (NGS)-based HLA typing has recently become more commonly used.]

HLA matching for the purpose of HSCT is generally confined to major class I and II loci, although it is increasingly appreciated that minor histocompatibility antigens also play important roles in the outcome of allogeneic HSCT. Any disparity at major HLA loci between the donor and the recipient requires vigorous immunologic intervention to avoid rejection, or, should engraftment occur, subsequent GVHD.

Donor selection

Any HLA-identical sibling, who does not have an anticipated risk for the underlying condition for which their sibling is undergoing transplant, should be considered a potential donor. ABO mismatch is not a contraindication, but if multiple donors are available, a donor with the same ABO type is preferred [major ABO mismatch (A or B or O) requires the removal of the red cells and/or plasma from the bone marrow graft prior to infusion]. In past studies, best outcomes have been associated with a fully matched HLA-matched sibling donor; however, only 20–25% of patients will have matched sibling donors within their family. When sibling donors are not available, volunteer unrelated donors who are HLA, A, B, C, and DRB1 compatible or minimally mismatched with the recipients are sought through a search of the international donor registries, including the National Marrow Donor Program.

An acceptable adult unrelated donor donating bone marrow or peripheral blood stem cells (PBSCs) should be matched via high-resolution DNA typing at least seven of eight alleles (HLA-A, B, C, and DRB1). When available, a higher degree of match (i.e., 10 of 10 alleles, including DQ, or 12 of 12, including DP, and matching as much as possible for HLADRB3/4/5 alleles) is desired.

Other alternatives include umbilical cord blood (UCB) donor, which has been matched via low-resolution typing of A and B with high-resolution DRB1 typing, at least four of six antigens (HLA-A, B, and DRB1), but better outcomes have been associated with matching at least six of eight antigens at the high-resolution typing level.

Suggested strategy for donor selection and prioritization

  • 1.

    Priority of donor by HLA disparity for a related donor is:

    • a.

      HLA-identical sibling

    • b.

      HLA-matched relative (fully matched 10/10)

    • c.

      Single HLA class I antigen or single class II allele mismatched relative (the class II antigen matching must be confirmed by high-resolution molecular typing)

  • 2.

    The priority of donor by HLA disparity for an unrelated, volunteer donor is:

    • a.

      HLA alleles matched at all loci, 10/10 match (A, B, C, DRB1, and DQB1)

    • b.

      If such a donor is not available, single-allele mismatch (7/8) at HLA A, B, C, and DRB1

      • i.

        Select an HLA-C*03:03 versus C*03:04 mismatch, if present (this mismatch is equivalent to a match)

      • ii.

        If possible, select a matched/permissive HLA-DPB1 mismatch based upon the algorithm developed by Crivello et al. ( https://www.ebi.ac.uk/ipd/imgt/hla/dpb_v2.html )

      • iii.

        Minimize mismatches at HLA DQB1 and DRB3/4/5

      • iv.

        Avoid mismatches targeted by HLA donor-specific antibodies (DSA) present in the recipient

  • 3.

    The priority for the selection of UCB units is:

    • a.

      High-resolution typing of HLA A, B, C, and DRB1 is best practice:

      • i.

        ≥4/8 High-resolution match between donor and recipient minimum

      • ii.

        Better outcomes with higher level of matching

    • b.

      Avoid mismatches in the donor UCB targeted by HLA DSA present in the recipient

    • c.

      Cell dose:

      • i.

        Total nucleated cell count (TNC)≥2.5×10 7 /kg, CD34≥1.5×10 5 /kg minimum single-unit dose, higher doses preferred (≥4×10 7 /kg)

      • ii.

        For 2-unit procedures, each should have TNC≥1.5×10 7 /kg, CD34≥1.0×10 5 /kg

Secondary donor prioritization (after HLA matching prioritization)

  • 1.

    HSC source : For recipients of HLA-matched related donor allografts, the preferred source of HSC (PBSCs), bone marrow, or UCB will be determined by the transplant protocol on which the recipient is enrolled, the disease status of the recipient, and/or the medical condition of the donor. Bone marrow is the preferred source for most allogeneic pediatric transplants. This is discussed later in more detail.

  • 2.

    Cell dose : When using bone marrow or peripheral blood as the HSC source, the donor’s size must be adequate to allow safe donation of the requested cell dose. This is more likely to be a consideration when the recipient is large-sized or in circumstances where bone marrow harvest is the preferred source of graft. While the cell dose can be altered when bone marrow or peripheral blood is the HSC source, UCB units have fixed cell doses.

  • 3.

    Donor gender : Male preferred, nonparous or single parous female next choice

  • 4.

    Donor age group : A younger donor is preferred unless the donor’s size would prohibit the collection of sufficient cells for transplantation.

  • 5.

    ABO compatibility : ABO match followed by minor mismatch followed by major mismatch and least preferred bidirectional mismatch.

  • 6.

    Donor parity : nonparous or lower parity preferred.

  • 7.

    Cytomegalovirus (CMV) serology: A CMV-negative donor is preferred for a CMV-negative recipient.

Table 30.6 lists the blood bank support required for HSCT.

Table 30.6
Blood bank support for hematopoietic stem cell transplantation.
Problem Solution
Transfusion-associated GVHD Standard: gamma radiation (2500–3000 cGy)
CMV transmission (CMV negative donor and recipient) Standard: use of CMV-negative blood products. Can also use CMV “equivalent” products that remove lymphocytes and leukocytes that can harbor viruses through filtration
Alloimmunization
  • 1.

    Graft failure or rejection (especially aplastic anemia)

Avoidance or minimizing of pretransplant transfusions; avoidance of transfusions from family members, especially potential donors
  • 2.

    Refractory thrombocytopenia a

  • 1.

    Most effective: leukocyte-depleted blood products

  • 2.

    Single-donor platelets

  • 3.

    HLA-matched donors

  • 4.

    Cross-matched platelets

ABO incompatibility
  • 1.

    ABO incompatibility

Recipient Donor
O A B AB
O Compatible Major Major Major
A Minor Compatible Bidirectional Major
B Minor Bidirectional Compatible Major
AB Minor Minor Minor Compatible

  • 2.

    Suggested processing for bone marrow grafts b

Donor blood group Recipient blood group
A B AB O
A None RBC and plasma depletion Plasma depletion RBC depletion
B RBC and plasma depletion None Plasma depletion RBC depletion
AB RBC depletion RBC depletion None RBC depletion
O Plasma depletion Plasma depletion Plasma depletion None

Abbreviations: CMV , Cytomegalovirus; GVHD , graft-versus-host disease; HLA , human leukocyte antigen; RBC , red blood cell; SOS , sinusoidal obstruction syndrome; VOD , veno-occlusive disease.

a Nonalloimmunization causes of refractory thrombocytopenia include drugs, hepatic VOD or SOS, hypersplenism, or sepsis. Another more unusual cause of refractory thrombocytopenia is a syndrome resembling thrombotic thrombocytopenic purpura, often associated with the use of Calcineurin inhibitors, total-body irradiation, or the development of acute GVHD.

b Isohemagglutinin titers in both donor and recipient may affect the specific requirements for processing with regard to need for RBC and/or plasma depletion.

HSC sources, collection, and manipulation

Bone marrow

Bone marrow harvesting is carried out using general or epidural anesthesia under sterile conditions.

The recommended cell dose and volume of marrow to be collected are determined by the recipient’s body weight and diagnosis, the type of graft manipulation (if any) that will occur, and the size of the donor.

The iliac crests (most common posterior and rarely anterior if an adequate number of cells are not obtained from posterior iliac crests) of the donor are prepared and draped. Approximately 5 mL of bone marrow is aspirated from each bone puncture until the required amount is obtained, with multiple bone punctures performed through a single skin puncture. The marrow is collected in a heparinized collection system. The minimum concentration of heparin (preservative-free) is 3–5 units/mL of bone marrow. The quantity of nucleated bone marrow cells required to ensure engraftment is 2–5×10 8 cells/kg of recipient body weight with a goal of >4–5×10 6 CD34+ cells/kg. The usual volume of marrow required to achieve this cell yield is 10–15 mL/kg of recipient body weight. The maximum amount one can take from a donor to avoid needing blood transfusion is 20 mL/kg donor weight. Marrow from children, especially infants, has a higher proportion of marrow-repopulating cells than marrow from older donors. The marrow is filtered through a filtering apparatus to remove bone and tissue fragments and placed in a blood transfer pack. In allogeneic transplants the bone marrow is then given to the recipient as an intravenous (IV) infusion over a period of a few hours.

Peripheral blood stem cells

PBSCs are the most commonly used source for an autologous graft, and bone marrow is reserved for cases where PBSC collection or mobilization is not feasible. PBSCs are collected from donors after stimulation with G-CSF (10 µg/kg per day) and have been successfully used to reconstitute hematopoiesis in recipients of autologous, syngeneic, and allogeneic grafts. Patients who receive >5×10 6 CD34+ cells/kg engraft satisfactorily (minimum 2×10 6 CD34+/kg required). In autologous donors a combination of myelosuppressive chemotherapy and G-CSF administration is more effective at mobilizing CD34+ cells. Plerixafor alone or in combination with G-CSF may be used in certain conditions for mobilization.

Plerixafor is usually used in patients or donors requiring PBSC collection who fail to mobilize, as defined by peripheral blood CD34+ cell counts <20 cells/µL following G-CSF-based mobilization protocols. Plerixafor used in children is off-label [not Food and Drug Administration (FDA) approved for use in children]. The standard dose is 0.24 mg/kg (maximum dose 40 mg) once daily subcutaneous for up to 4 consecutive days. Each dose should be given around 10 hours prior to apheresis. The use of high-dose Plerixafor 0.48 mg/kg once daily, to improve mobilization, has also been reported.

Umbilical cord blood

UCB has been collected, cryopreserved, and is used as the source of pluripotential HSCs when a related or unrelated stem cell donor is not available. UCB cells have increased proliferative capacity and decreased alloreactivity, with a lower incidence of GVHD, in addition to immediate availability. These properties, coupled with an absence of donor risk, is an advantage in the use of this source of HSC, but disadvantages include the inability to fully evaluate the genetic history of these donors, limited cell dose, and lack of feasibility of donor lymphocytes infusion if needed. For a single UCB transplant, the standard minimum cell dose must be equal to or >2.5×10 7 TNC/kg of recipient weight. If the transplant is performed with two cord blood units, the preferred dose of the combined units is ≥3.0×10 7 TNC/kg of recipient weight. A higher cell dose is generally preferred and especially when there is a greater degree of HLA mismatch between the cord unit(s) and the intended recipient. For cord blood the interaction between the cell dose available and HLA matching affects the selection strategy as follows:

  • 1.

    Single unit:

    • a.

      The UCB unit is a 6/6 match with the recipient with a cell dose ≥2.5×10 7 TNC/kg.

    • b.

      The UCB unit is a 5/6 matched with a cell dose ≥4.0×10 7 TNC/kg.

    • c.

      The UCB unit is a 4/6 matched with a cell dose ≥5.0×10 7 TNC/kg.

  • 2.

    Double units should be adequately matched with each other and the recipient as discussed earlier.

Graft manipulation postcollection

Several manipulations can be performed on HSC postcollection to reduce the risk of red blood cell (RBC) hemolysis due to ABO incompatibility, GVHD, and the reinfusion of malignant cells.

This is in addition to CD34 selection or other methods to remove T cells [e.g., T-cell receptor (TCR) alpha/beta-positive T-cell depletion] which are often performed, on experimental protocols, to overcome HLA mismatch between the donor and the recipient, especially when using HLA-haploidentical donors.

ABO incompatibility

ABO incompatibility between donor and recipient is encountered in 25–30% of allogeneic transplants. A major incompatibility occurs when the recipient plasma contains isohemagglutinins directed against the donor RBC antigens (e.g., group O recipient, group A donor), and minor incompatibility occurs when the donor plasma contains isohemagglutinins directed against recipient RBC antigens (e.g., group A recipient, group O donor). Red cell depletion is required for major mismatches in order to minimize the amount of incompatible blood in the graft. After the transplant procedure, patients can be followed with immunohematologic testing for the appearance of donor-derived RBCs and changes in recipient isohemagglutinin titers.

Graft-versus-host disease

Activation of allogeneic T cells against recipient antigens is termed GVHD. All forms of allogeneic HSCT require approaches to reduce the risk of GVHD, most commonly in the form of GVHD prophylaxis using medications such as cyclosporine or tacrolimus. Reduction of T lymphocytes from allogeneic HSC postcollection decreases the risk of GVHD and is sometimes part of the approach to GVHD prevention. This can be achieved through various techniques, including monoclonal antibodies accompanied by complement-mediated lysis, immunotoxins, or immunomagnetic beads (complete or partial in vivo T-cell depletion may include administration of ATG or alemtuzumab). An alternative method is to administer post-HSCT cyclophosphamide to accomplish in vivo T-cell depletion following infusion of an unmanipulated graft. This approach is commonly used for HLA-haploidentical donor grafts but is increasingly being used for other graft types as well.

Purging of malignant cells

Purging can be performed to remove malignant cells. This can be achieved through the use of monoclonal antibodies combined with complement, monoclonal antibodies linked to toxins or various chemotherapy drugs. This has been used in studies of autologous HSCT for neuroblastoma, and published data from a phase 3 randomized trial showed that purging of PBSC did not improve outcome. Because of this and other studies, purging tumor from autologous PBSC is not commonly practiced.

Medical evaluation of HSC donors

Donor evaluation procedures protect the safety of the HSC donor and recipient. A comprehensive evaluation of donors seeks to identify potential medical risks to both the donor and the recipient.

The medical history should focus on indicators of active disease processes and the risk of an indolent infectious disease. The history should include details of vaccination, travel, blood transfusions, and questions to identify persons at high risk for the transmission of communicable or inherited, hematological or immunological diseases, and questions to identify any past history of malignant disease.

Donor assessment should also include pregnancy testing, prior deferrals from blood donation, contraindications to blood donation, and findings that would increase the anesthesia risk (an electrocardiogram and chest X-ray are commonly performed on adult donors). PBSC donors should be evaluated for potential contraindications to central venous access catheter placement and G-CSF for HSC mobilization.

Laboratory evaluation for potential donors should include:

  • Complete blood count

  • Comprehensive metabolic panel (including electrolytes, glucose, blood urea nitrogen and creatinine, serum protein, serum albumin, aspartate aminotransferase (AST), and alanine aminotransferase (ALT))

  • ABO group and Rh type, red cell antibody screen

  • Confirmatory HLA typing

  • Testing for infectious diseases, including:

    • Human immunodeficiency virus (HIV), type 1 (HIV-1)

    • HIV-2

    • Hepatitis B virus

    • Hepatitis C virus

    • Treponema pallidum (syphilis)

    • Human T-cell lymphotrophic virus I (HTLV-1)

    • HTLV-2

    • CMV

    • Epstein–Barr virus

    • Herpes viruses

Additional testing may be recommended based on local regulations or as clinically indicated [e.g., West Nile virus, Trypanosoma cruzi (Chagas’ disease), screening for hemoglobin S, SARS-CoV-2].

Table 30.7 lists the pretransplantation evaluation of the HSCT recipient.

Table 30.7
Pretransplantation evaluation of the recipient a .
History, physical examination, weight, height, BSA, and head circumference (if appropriate for age)
Complete blood count
Comprehensive metabolic panel
24-h urine for creatinine clearance or GFR
ABO blood group, Rh typing, and isohemagglutinin titer (if indicated depending on donor blood group)
HIV-1 and -2, HTLV I and II b
CMV, hepatitis B and C, EBV, and HSV 1/2 b
Syphilis b
Echocardiogram (or MUGA) with ejection fraction (or shortening fraction) as appropriate and EKG
PFT (if age appropriate and feasible) or O 2 % saturation
Imaging: Chest X-ray at minimum (additional imaging requirements vary depending on the specific transplant program) c
Dental and eye evaluation
Disease-specific manifestation assessments at baseline as clinically indicated (e.g., bone marrow aspiration/biopsy if applicable)
Karnofsky or Lansky score (age appropriate)
Psychosocial evaluation
Abbreviations: BSA , Body surface area; CMV , cytomegalovirus; CNS, central nervous system; CT , computed tomography; CVL , central venous line; EBV , Epstein–Barr virus; GFR , glomerular filtration rate; HIV , human immunodeficiency virus; HSCT, hematopoietic stem cell transplantation; HSV , herpes simplex virus; HTLV , human T-cell lymphotrophic virus; MRI , magnetic resonance imaging; MUGA, multigated acquisition scan; PFT , pulmonary function test.

a Additional testing may be needed as indicated to evaluate any existing comorbidities.

b Antigen detection or molecular methods may need to be used for testing especially if primary disease involves immune deficiency. All HSCT recipients require insertion of CVL, the specific CVL type used is different depending on patient, disease, transplant center, preference, and availability.

c CT scan chest, abdomen, and pelvis and CT of the neck if clinically warranted to rule out infection; MRI of the brain for all to establish a baseline for future evaluation of possible CNS toxicities may be done based on the specific transplant program standards.

Pretransplantation preparative regimens (conditioning)

Pretransplantation conditioning is used both to eradicate disease and as a means of immunosuppressing the recipient sufficiently to allow the acceptance of a new immunologically disparate hematopoietic system. Conditioning regimens may vary between centers and research protocols. Table 30.8 shows examples of preparative conditioning regimens. The current mainstay preparatory regimen for traditional myeloablative HSCT consists of ablative doses of either total body irradiation (TBI) or busulfan (Bu) together with either one or two chemotherapy agents. On completion of the preparative regimen, the donor marrow, PBSC, or UCB is infused.

Table 30.8
Commonly used stem cell transplantation preparative regimens.
Cyclophosphamide total dose Additional drug Additional drug total dose Total body irradiation (cGy) (fractionated)
120 mg/kg 1200–1350
Etoposide 50–60 mg/kg 1200–1350
120 mg/kg Etoposide 30 mg/kg 1200
120–200 mg/kg Busulfan a 16 mg/kg
Melphalan 140 mg/m 2 1200–1350

a Busulfan is commonly administered as IV medication and is usually dosed initially as follows: weight <10 kg: 0.8 mg/kg/dose; weight ≥10 kg and age <4 years: 1 mg/kg/dose; age ≥4 years: 0.8 mg/kg/dose. Subsequent doses are usually adjusted to achieve the target of (AUC) Area Under the Curve 900–1500 μmol/L per minute (or net steady-state concentration (SSc) of 800–1200 ng/mL) (target can be slightly variable depending on local center and treatment protocol).

Commonly used conditioning regimens

Leukemia

Two conditioning regimens (shown in Table 30.8 ), which have antitumor as well as immunosuppressive properties, are commonly used prior to stem cell transplantation. TBI (fractionated doses) and cyclophosphamide (or etoposide) are utilized in acute lymphoblastic leukemia (ALL). A combination of TBI and cyclophosphamide or Bu and cyclophosphamide is commonly used for acute myeloid leukemia (AML), chronic myeloid leukemia (CML), myelodysplasia, and juvenile myelomonocytic leukemia.

Non-Hodgkin lymphoma

Myeloablative chemotherapy regimens such as cyclophosphamide, bis-chloroethyl-nitroso-urea (BCNU) or carmustine, etoposide (VP-16) (CBV) ( Table 30.9 ) or BCNU, etoposide, cytosine arabinoside, and melphalan ( Table 30.10 ) are conditioning regimens commonly utilized prior to stem cell transplantation in relapsed non-Hodgkin lymphoma (NHL).

Table 30.9
CBV regimen a for stem cell transplantation in non-Hodgkin lymphoma.
Days Chemotherapy
−8, −7, −6 BCNU: 100 mg/m 2 per day IV over 3 h (total dose 300 mg/m 2 )
Etoposide 800 mg/m 2 per day IV as a 72-h continuous infusion (total dose 2400 mg/m 2 )
−5, −4, −3, −2 Cyclophosphamide: 1500 mg/m 2 per day IV over 1 h. Use MESNA (total dose 2200 mg/m 2 IV every 24 h)
−1 No treatment
0 Stem cell infusion
Notes : Methylprednisolone 1 mg/kg per day in divided doses given every 6 h for pulmonary protection from BCNU toxicity on days −9 to −2. Wean by 20% every 2 days, taper.
Abbreviations: BCNU , Bis-chloroethyl-nitroso-urea; IV , intravenous.

a Various dosing schedules for CBV regimen exist.

Table 30.10
Bis-chloroethyl-nitroso-urea (BCNU), etoposide, cytosine arabinoside, and melphalan regimen for stem cell transplantation in non-Hodgkin lymphoma.
Day Chemotherapy
−6 BCNU: 200–300 mg/m 2 IV over 3 h
−5, −4, −3, −2 Etoposide 200 mg/m 2 per day IV over 1 h (total dose 800 mg/m 2 )
−5, −4, −3, −2 Cytosine arabinoside: 400 mg/m 2 per day IV over 1 h or 200 mg/m 2 q12h for eight doses (total dose 1600 mg/m 2 )
−1 Melphalan 140 mg/m 2 IV over 30 min
0 Stem cell infusion
Notes : Methylprednisolone 1 mg/kg per day in divided doses given every 6 h for pulmonary protection from higher dose BCNU toxicity on days -7 to -2. Wean by 20% every 2 days for an 8-day taper.
Abbreviation: IV , Intravenous.

Solid tumors

HDC with autologous stem cell rescue has become the standard of care for neuroblastoma and medulloblastoma (especially in infants and younger children to spare radiation use) and has been utilized in the setting of other relapsed solid tumors.

Severe aplastic anemia

Patients receiving matched sibling HSCT for severe aplastic anemia (SAA) have traditionally received cyclophosphamide 50 mg/kg per day for 4 days, along with equine ATG 30 mg/kg daily during 3 of the cyclophosphamide days. European approaches have successfully given lower doses of cyclophosphamide by adding several doses of fludarabine. For unrelated donor HSCT for SAA, regimens containing rabbit ATG, fludarabine, cyclophosphamide, and low-dose TBI have led to excellent outcomes in patients with fully matched or single-antigen mismatched unrelated donors. Table 30.11 describes a commonly used immunosuppressive HSCT preparative regimen for SAA. Long-term survival in the range of >90% can be expected with HSCT using histocompatible related donors. The overall risk of unrelated or mismatched related donor transplantation precludes its use as front-line therapy for SAA at this time, although this is an area of ongoing study. As improved HLA typing, preparative regimens, and GVHD prophylaxis are utilized, HSCT is becoming available to a wider group of patients with SAA and is being evaluated in prospective clinical trials, using unrelated and haplo donors.

Table 30.11
Hematopoietic stem cell transplantation preparative regimen for severe aplastic anemia.
The following regimen is commonly used for nonsensitized (not heavily transfused) matched sibling donors a :
Day−5 Morning: Cyclophosphamide, 50 mg/kg IV over 1 h
Afternoon: eATG b , 30 mg/kg IV. First dose of ATG is given over 8 h; subsequent doses are given over 4 h
Day−4 (same as for Day−5)
Day−3 (same as for Day−5)
Day−2 Morning: Cyclophosphamide, 50 mg/kg IV over 1 h
Day−1 Rest; cyclosporine A, 10 mg/kg per day PO daily adjusted for serum levels
Day−0 Marrow infusion
Abbreviations: eATG , Equine antithymocyte globulin; IV , intravenous; PO, Per os; rATG ; rabbit antithymocyte globulin; TBI , total body irradiation.

a The following regimen is commonly used for sensitized (heavily transfused) matched sibling, or unrelated donors: rATG 3 mg/kg Day −4 to −2, Fludarabine 30 mg/m 2 Day−5 to −2 and TBI 200 cGy Day−1. Cyclophosphamide 50 mg/kg per day may be added for 1–2 days per transplant center local standard.

b eATG may be substituted by rATG 3 mg/kg per day per center local standard.

Fanconi anemia

Due to impaired tissue repair and increased risks of adverse effects, reduced-intensity conditioning regimens are employed.

Matched sibling graft:

  • Cyclophosphamide 10 mg/kg per day×4 days

  • Fludarabine 35 mg/m 2 per day×4 days

  • ATG (rabbit) 2.5 mg/kg per day×4 days

Unrelated bone marrow graft:

  • TBI 300 cGy

  • Cyclophosphamide 10 mg/kg per day×4 days

  • Fludarabine 35 mg/m 2 per day×4 days

  • ATG (rabbit) 2.5 mg/kg per day×4 days

Cord graft:

  • Similar to unrelated donor marrow or alternatively

  • Bu 1 mg/kg×2 doses/day×2 days

  • Cyclophosphamide 10 mg/kg per day×4 days

  • Fludarabine 35 mg/m 2 per day×4 days

  • ATG (rabbit) 3 mg/kg per day×4 days

  • Methylprednisolone 2 mg/kg per day, though +20 then taper

Miscellaneous conditions

In hemoglobinopathies, selected immunodeficiency, and other inherited disorders, a commonly used myeloablative conditioning regimen is Bu and cyclophosphamide with or without ATG. Many centers are using reduced toxicity/reduced intensity approaches for these patients.

You're Reading a Preview

Become a Clinical Tree membership for Full access and enjoy Unlimited articles

Become membership

If you are a member. Log in here