Graft Engineering and Cell Processing


It has been more than 70 years since allogeneic hematopoietic stem cells (HSC) were first transplanted to human recipients in the form of bone marrow. Since that time, the source of HSC has widened to include autologous bone marrow, growth factor mobilized peripheral blood progenitor cells (PBPCs), umbilical cord blood, and more. In parallel, there has been the development of other cellular therapies to treat a variety of diseases (such as the use of T cells genetically modified to express a chimeric antigen receptor (CAR) for the treatment of leukemias) and mesenchymal stromal cells (MSCs) to treat or prevent graft-versus-host disease (GVHD). This chapter provides a general overview of how these cells are prepared ex vivo, tested, and released for clinical use. Preparation procedures range from the simple depletion of plasma or incompatible erythrocytes from HSC grafts to complex genetic modifications of other cells using viral vectors to induce tumor-associated antigen specificity.

The rapid development of cellular administrations for therapeutic applications has attracted the interest of regulatory authorities (e.g., the Food and Drug Administration [FDA] in the United States). Understanding the relevant regulations is critically important when undertaking the manufacture of these types of products.

Regulatory Issues With Cell Processing

In response to the increase in the use of therapeutic cell products, the FDA, and other national regulatory authorities, have had to develop a relevant regulatory strategy to cover these products. The approach used by the FDA, and some other agencies, has been to develop a risk-based method ( Table 103.1 ). This is based on an assessment of the relative risks posed broadly by the collection of the initial cells, their ex vivo manipulation and storage, and their administration to the recipient. Good Manufacturing Practices (GMP) regulations have existed for some time and are applied in the drug field, primarily in the preparation of small molecules. This was extended to certain cellular therapy products when these were classified as drugs for regulatory purposes. The applicable GMP regulations appear in Title 21 of the Code of Federal Regulations, primarily in Parts 210/211. They cover all aspects of manufacturing, testing, packaging, storing, releasing, and distributing drugs. Recognizing that these regulations were probably overkill for experimental cellular therapeutics being developed by an academic institution, the FDA published a guide indicating the level of GMP compliance required for products to be used in Phase 1 clinical trials. Simultaneously, the Agency recognized that there were certain products whose preparation and use posed a substantially lower risk (e.g., PBPC) since their collection was straightforward, their ex vivo manipulation was simple and their clinical use did not result in adverse events, and was of clinical benefit. As a result, in 2005 they published Good Tissue Practice (GTP) regulations as a part of 21 CFR Part 1271 (Human Cells, Tissues, and Cellular and Tissue-Based Products [HCT/Ps]) to cover the preparation of these products. These were established nominally to prevent the introduction, transmission, and spread of communicable diseases by HCT/Ps. This was based on the presumption that most post-transplant infections and admissions to intensive care units were attributable to the administration of contaminated products. The validity of this assumption is open to question. GTP regulations provide a framework for screening, performing ex vivo processing, storing and distributing HCT/Ps, and providing the FDA with an overview of current activities by requiring an annual registration of collection and processing facilities. The Part 1271 regulations effectively filled a gap in the law that had left unregulated HCT/Ps that were minimally manipulated (e.g., were not cultured ex vivo, genetically modified, activated ex vivo), intended for homologous use, or not combined with another article (e.g., a matrix or scaffold) for administration. Minimally manipulated HCT/Ps should not exert a systemic effect and should not be dependent on the metabolic activity of living cells for them to function. If this was not the case, then the cells should be for autologous use only, for use in a first- or second-degree blood relative, or in reproductive use. Cellular products that fall into this classification are Type 361 products. The Part 1271 regulations do not apply to vascularized organs for transplantation, whole blood, blood components, or minimally manipulated BM for homologous use that is not combined with another article. This means that traditional marrow-derived hematopoietic stem cell transplants (HSCT) do not fall under FDA regulations and that these procedures are currently considered a practice of medicine. Products that fall under IND regulations are referred to as Type 351 products. These are cells that have been cultured ex vivo and transduced or activated ex vivo and, therefore, are more-than-minimally manipulated. The facility processing these cells is required to operate under GMP. These regulations were developed to ensure that products are prepared under a controlled and auditable process that ensures their safety, purity, and potency. As indicated previously, the agency has recognized the differences between products prepared for a phase 1 clinical trial and, therefore, the application of GMP to cellular therapy products follows a continuum, such that as the trial proceeds, the expectation is that implementation of GMP will become more rigorous, such that a phase III product would be extensively characterized and manufactured using a fully validated process.

Table 103.1
Characteristics of Products Regulated Under Good Tissue Practices and Good Manufacturing Practice Regulations and Associated Guidance Documents
Good Tissue Practices (gtp) (Type 361 Products) 21cfr Part 1271 Good Manufacturing Practices (gmp) (Type 351 Products) 21cfr Parts 210 & 211 Food and Drug Administration (fda) Guidance
Minimally-Manipulated More-than-Minimally Manipulated
  • Regulatory Considerations for Human Cells, Tissues, and Cellular and Tissue-Based Products: Minimal Manipulation and Homologous Use.

  • December 2017

Homologous use only Includes non-homologous use
No combination products Combination (e.g., with matrix or scaffold) products Current Good Manufacturing Practice Requirements for Combination Products. January 2017Requesting FDA Feedback on Combination Products. December 2019
No systemic effects unless for:

  • Autologous use

  • Allogeneic use in 1st- or 2nd-degree relative

  • Reproductive use

Systemic effects
Donor Eligibility AssessmentRegister establishment with FDA annually
  • Investigational New Drug/Device Application required

  • Annual IND Report

  • Eligibility Determination for Donors of Human Cells, Tissues, and Cellular & Tissue-Based Products (GTP)

  • August 2007.

  • Investigational New Drug Applications Prepared and Submitted by Sponsor-Investigators (GMP)

  • May 2015

  • IND Applications for Clinical Investigations: Regulatory and Administrative Components (GMP)

  • February 2018

Implementation of Part 1271 regulations has impacted the “routine” laboratory that prepares cells primarily for HSCT. Laboratories that use cells other than bone marrow (BM) now must register annually with the FDA; ensure that donors meet eligibility requirements (or document why non-eligible donors are used); and manufacture, store, and distribute the cells under GTP. If a laboratory has previous experience manufacturing products under GMP conditions, it will already be familiar with most of the features of GTP. Generally, these cover personnel, procedures, facilities, environmental control and monitoring, equipment, supplies and reagents, recovery, processing and process controls, process changes, process validation, labeling controls, storage, receipt, pre-distribution shipment and distribution, records, tracking, and complaints. Implementation of the components of GMP and GTP operations is a time-consuming process that requires the development, implementation, and maintenance of numerous components and generates a considerable volume of documentation. Professional societies, such as the Foundation for the Accreditation of Cellular Therapy (FACT) and AABB (formerly the American Association of Blood Banks), have developed standards and an accreditation process that takes into account GTP/GMP regulations and provides a framework around which compliance can be built.

In the United States, cord blood is a licensed product, and facilities that prepare and bank cord blood are required to obtain a Biologics License. This allows the manufacturer to introduce, or deliver for introduction, a biologic product into interstate commerce (21 CFR 601.2). There are professional standards that address cord blood collection, manufacturing, and use.

Central to both GMP and GTP regulations is the establishment and maintenance of a quality program. This quality program must ensure that the appropriate regulations are being followed on an ongoing basis, that mechanisms are in place for detecting, reviewing, and remediating errors and deviations from regulations, policies, and procedures, and that an audit program will be developed, implemented, and maintained. Activities performed by the quality program must be documented, and the program should be staffed by individuals who are not involved in the hands-on manufacturing of products.

Holders of INDs must provide the agency with an annual report on the protocol, including a listing of the products administered and those that have been prepared but not used. In addition, cell processing facilities should be prepared to assist the IND sponsor by providing information on products that have been associated with severe adverse reactions in the recipients and whether these were attributable to product quality. For Type 361 products, the facility must, as Biological Product Deviations, report any contaminated products that have been administered to a patient.

Professional Standards

The two major accrediting organizations in the United States for cellular therapies are FACT and AABB. Whereas FACT offers accreditation for collection, processing, and clinical use of cellular therapy products, the AABB focuses on collection and laboratory processing. Both organizations inspect based on standards that are published every 18 months to 3 years. In the case of FACT, the standards are published in collaboration with the Joint Committee on Accreditation in Europe (JACIE). FACT also publishes separate standards that cover cord blood banking (in collaboration with NetCord) and for Immune Effector Cells. Both organizations have worked to harmonize their standards with American, Canadian, Australasian, and European regulatory agencies; therefore, accreditation by either organization is of great assistance on the pathway to regulatory compliance. Several other professional organizations accredit particular aspects of operations within the cell processing facility. These include the College of American Pathologists (CAP), which accredits general laboratories and hematology and flow cytometry facilities, the American Society for Histocompatibility and Immunogenetics (ASHI), and the European Federation for Immunogenetics (EFI), which accredit histocompatibility testing laboratories. Some organizations, such as CAP and StemCell Technologies, also provide proficiency testing services for laboratory staff.

Manipulation of Hematopoietic Stem Cell Transplantation Products

Hematopoietic grafts are manipulated in vivo to remove an unwanted component that may cause adverse effects or to enrich the desired population, such as HSC. As discussed previously, the degree of manipulation may determine the regulations under which the product is manufactured and handled. The FDA defines minimal manipulation as processing that does not alter the relevant biologic characteristics of the cells or tissues. This includes procedures such as RBC, plasma depletion, and cell selection using an approved device. By contrast, more-than-minimal manipulation would include activities such as culture ex vivo, genetic modification, and ex vivo activation. As stated previously, manufacturers of minimally manipulated products would follow GTP regulations, whereas products that are more-than-minimally manipulated would fall under GMP regulations.

Routine Minimal Manipulation for Volume Reduction or ABO Incompatibility

The most widely used form of manipulation in the hematopoietic progenitor cell (HPC) processing facility probably is the removal of erythrocytes and plasma to overcome incompatibility between donor and recipient ( Table 103.2 ). This process is performed using techniques that were developed by the blood banking industry.

Table 103.2
Processing Performed to Address ABO Incompatibilities Between Hematopoietic Stem Cell Donors and Recipients
Recipient ABO Type Donor ABO Type Type of Processing
ABO identical ABO identical No special processing required
A or B AB RBC depletion
O A/B/AB RBC depletion
A B RBC + plasma depletion
B A RBC + plasma depletion
Antibody to RBCs N/A RBC depletion
A/B/AB O Plasma depletion
AB A or B Plasma depletion
N/A , Not applicable; RBC , red blood cell.

Plasma depletion to remove donor antibodies that may react with recipient cells is achieved by centrifugation of the graft, usually in a transfer pack, at approximately 2000 g for 10 minutes at ambient temperature. The pack is then placed in a plasma expresser, which compresses the product bag so that plasma can be forced out and into a separate collection bag. Plasma depletion can also be used to reduce the volume of ABO-compatible grafts when the donor is larger than the recipient.

Erythrocyte depletion removes incompatible donor erythrocytes that would stimulate a reaction by the recipient upon administration. Most facilities establish a maximum volume of incompatible RBCs that can be infused with HPC; exceeding this limit can result in hemolysis and a transfusion reaction. Depletion of erythrocytes can be achieved most simply by centrifugation at approximately 3000 g for approximately 10 minutes at ambient temperature. The leukocyte-rich buffy coat is then collected from the interface between the plasma layer and the RBCs.

Erythrocyte depletion can also be achieved by sedimenting erythrocytes using hydroxyethyl starch (Hetastarch). This promotes RBC sedimentation by the formation of erythrocyte rouleaux. The hematocrit of the product is first adjusted to ∼25% by the addition of normal saline, and 6% Hetastarch (Hespan) is added at a volume:volume ratio of 1:6 to 1:7. Sedimentation can be performed under gravity or accelerated by centrifugation.

A more rigorous erythrocyte depletion is achieved by centrifugation of the collection on a Ficoll-Hypaque density gradient. The cells are layered onto a density gradient cushion of Ficoll-Hypaque and centrifuged at 400 g for approximately 30 minutes. The mononuclear cells collect at the interface and are removed and washed before use. This process depletes erythrocytes, platelets, and granulocytes. The overall nucleated cell recovery is lower than with other techniques in which granulocytes are retained.

Automated devices are available for preparing buffy coats and density gradient-enriched cells. For larger volumes, the COBE 2991 Cell Processor from Terumo can be used. This requires a minimum volume of 150 mL RBCs for operation and may therefore not be suitable for pediatric processing. It is capable of preparing buffy coats and density-separated cell preparations using a functionally closed disposable set.

The COBE Spectra from Terumo is commonly used to collect PBPC by apheresis. The device is also less widely used in the processing facility to enrich mononuclear cells from BM.

For smaller starting volumes, the Sepax device from Cytiva (formerly GE Cell Therapy) ( Fig. 103.1A ) can be used. It has found widespread application in cord blood banks for buffy coat preparation (with or without the addition of hydroxyethyl starch) and has also been used for volume reduction of PBPC collections, density gradient separation of BM, and cell washing. The device has a small footprint, uses functionally closed disposables, and provides a print-out of operations.

Figure 103.1, AUTOMATED CELL SEPARATION AND CULTURE DEVICES.

The AXP II System for cord blood processing is available from Thermogenesis. The device is designed for enriching mononuclear cells from cord blood that are transferred to the processing set, which is then placed into the AXP device. This fits into a centrifuge bucket, and during spinning, the red and mononuclear cells are collected into separate bags, and the plasma is retained in the processing set. The PXP System performs a similar procedure on marrow harvests. Both devices provide closed sterile systems, and each system comes with software that enables data tracking to assist with regulatory compliance.

Purging of Autologous Grafts

Autologous HPC can be used for recipients lacking a human leukocyte antigen (HLA)-matched related or unrelated donor. It has been proposed that occult viable tumor cells collected with the graft and returned to the patient could act as a source for disease relapse. Gene marking studies have supported this hypothesis. As a result, much effort has been exerted to develop methods for the ex vivo detection and removal of tumor cells from autologous grafts ( Table 103.3 ). Techniques have included incubation with chemotherapeutic drugs, such as 4-hydroperoxy cyclophosphamide (4-HC), photosensitizing agents, and antisense oligonucleotides. Alternatively, tumor-directed monoclonal antibodies (MAb) can be used to identify the cells and effect their removal. The MAb-coated tumor cells can be eliminated by the addition of serum complement or by capturing them on a solid phase, such as a column matrix, a plastic sheet, or magnetic particles. These particles may be large (5 μm diameter—Dynabeads, Invitrogen), so they can be collected with the attached tumor cells in a standard magnetic field. The matrix material may be much smaller, such as iron-dextran nanoparticles (Miltenyi Biotec Bergisch Gladbach, Germany) or ferrofluids (Ferrotec Santa Clara, CA), which coat the cells. These are then collected on a metal matrix placed in a field generated by permanent magnets. Such systems are capable of depleting 4 to 6 logs of tumor cells from a graft. However, even at such high efficiencies, and given the limits of our ability to detect residual tumor cells, the clinical value of purging autologous grafts is debatable. There has also been a decline in interest in purging techniques because of the potential benefits of a graft-versus-tumor (GVT) effect detected in recipients of allogeneic grafts.

Table 103.3
Methods Used for Manipulation of Hematopoietic Grafts
Destruction in Situ Physical Separation
Tumor: Cytotoxic drugs (e.g., 4-HC a ) T cells: Rosetting with sheep erythrocytesLectins (e.g., soybean agglutinin)Centrifugal elutriation
Monoclonal Antibody-Based
  • Tumor or T cells:

  • Antibody + Complement

  • Immunotoxins (e.g., ricin)

  • Panning and immunoaffinity columns

Immunomagnetic separation

  • T cells: Negative selection (Pan T-cell and T-cell subset removal)

  • Tumor: Negative selection (using tumor-associated antigens)

  • Positive selection (Tumor and T cells) (CD34 enrichment) CliniMACS device

a 4-Hydroperoxycyclophosphamide.

T-Cell Depletion of Allogeneic Products

T cells in HPC grafts have the potential to cause severe or lethal GVHD or exert a beneficial GVT effect. Considerable work has been done to determine whether or not these opposing effects are produced by distinct subpopulations of T lymphocytes. This would allow ex vivo manipulation of allogeneic grafts to remove differentially the GVHD-producing T cells while sparing those that mediate GVT responses. Various subpopulations of T cells have been identified as candidate effector subpopulations; however, there is still no widespread consensus as to which subsets should be targeted.

Methods are available for Pan-T cell elimination from grafts using approaches similar to those used for purging tumor cells (see Table 103.3 ). Early techniques included the use of soybean agglutinin to aggregate the majority of non-progenitor cells and the rosetting of sheep erythrocytes with T cells to facilitate their removal. Although successful, these techniques are not “FDA friendly” and do not offer the specificity that likely is required to engineer T-cell subpopulations in allogeneic grafts. This is possible by using MAb directed toward the antigens that currently are used to identify T-lymphocyte subpopulations. The target population then can be removed with high efficiency using immunomagnetic separation, as described previously for purging autologous grafts. The challenge remains to identify the appropriate target T cell populations and to source clinical-grade MAb for these procedures. Several potential target antigens have been identified and separation techniques implemented. They range from pan–T-cell depletions using MAb to CD3 and CD2, depletions of helper and cytotoxic T cells using MAb against CD4 and CD8, to stimulation and removal of alloreactive populations by targeting activation antigens. In addition, methods have been developed for the selective manipulation of natural killer (NK) cells, natural killer T (NKT) cells, myeloid suppressor populations, and MSCs for administration to stem cell transplant recipients to prevent or treat GVHD.

Efforts have recently focused on the depletion of αβ-positive T cells from the graft. This spares the donor-derived alloreactive NK and γδ T cells, which may provide a tumor-directed response. Promising early clinical results have been obtained using this approach. Twelve patients received marrow or PBPC grafts depleted of ∼4 logs of αβ T cells, and all were alive at 3 months, and after 1 year, two had died. There was no severe GVHD (grade III to IV) post-infusion. A systematic review of the impact of γδ cells on the outcome of transplants concluded from 11 studies (919 patients), with a median follow-up of 30 months, that high γδ numbers were associated with lower relapse, fewer viral infections, and higher overall and disease-free survivals; however, no association was found between high γδ numbers and GVHD incidence.

Regulatory T Cells

A different approach for GVHD is to administer T reg cells post-transplant to prevent and treat GVHD. A first-in-man clinical study was performed using in vitro expanded T reg from partially HLA-matched third-party umbilical cord blood units that were administered to 23 patients receiving double cord blood transplants. Results were compared to 108 historical controls. No T reg acute toxicities were seen, and there was a reduced incidence of grades II-IV acute GVHD but since this was a Phase 1 study, it was not designed to demonstrate efficacy. Later studies support the finding that donor T reg infusions early after transplant can prevent GVHD after allogeneic transplantation. There are several questions still to be addressed. These include the source of T reg cells (cord versus peripheral blood—cord blood generally yields cells of a higher purity but lower numbers); whether the cells should be expanded in vitro before use; the best method for their purification; and the best conditions for expansion.

T reg represents a rare cell population whose phenotype cannot be categorically defined. They are generally enriched using their expression of CD25, sometimes incorporating prior depletion of C8 and CD19-positive cells. This leads to an enrichment of CD4 + CD25 + FOXP3 + cells which can be further increased to 87% purity by further in vitro expansion. The optimal method for enrichment appears to be the separation of CD4 + CD25 hi CD127 lo cells. Peripheral blood T reg cells can be effectively expanded in vitro by stimulation with CD3 and CD28 MAb and high concentrations of IL-2. Alternatively, artificial antigen-presenting cells (APCs) primed with CD3 (and CD28) MAb have been shown to preserve FOXP3 expression while achieving cell expansion. Expansion of cord blood T re can be further improved by using APC additionally primed with OX40L or 4-IBBL. Rapamycin can also be used to enhance TGF-β-dependent FOXP3 expression and to limit the activation and expression of effector T cells. It should be noted that cryopreservation of T reg may impair their function.

Positive Selection of Progenitor Cells

Methods that eliminate or physically remove either T cells or tumor cells from grafts are referred to as negative selection techniques . They are affected by variables such as target antigen expression, the sensitivity of detection technologies for quantitating separation efficiency, and other technical hurdles. These may be difficult to control in order to achieve the ideal composition of the graft, and the target level of residual T cells, or T cell subsets, remains to be established. A dose of 10 5 T cells/kg is generally regarded as the goal to minimize the risk of GVHD while facilitating engraftment. There are also no approved devices for negative selection, so these types of procedures must be performed under an IND.

For many years, the goal was to replace the negative selection with a procedure in which HPC populations could be enriched by positive selection. This would effectively deplete T cells, or tumor cells, from allogeneic and autologous grafts, respectively. The problem was the lack of a method for identifying the target HSC until the CD34 antigen was identified on a small population of progenitor cells, including the pluripotent cells required for hematopoietic transplantation. The subsequent availability of MAb directed against this antigen made possible the development of techniques for the enrichment of these cells. Immobilization of the antibodies on a matrix (e.g., plastic sheets),cellulose, and magnetic particles were used as the primary approach, and several devices were commercially developed (see Table 103.3 ). The first to achieve FDA approval for use with apheresis products were the Baxter Isolex 300i, which used Dynal 5 μm magnetic beads as the separation modality and released CD34 + cells from the beads using a competitive binding peptide. This device was taken over by Nexell, which ultimately went out of business in 2002.

The alternative is the CliniMACS system (Miltenyi Biotec, Fig. 103.1C ), which currently has regulatory approval in Europe, but still requires an IND for use in the United States for other than one specific indication (the depletion of T cells by CD34 + cells selection for the treatment of acute myeloid leukemia). This uses anti-CD34 or anti-CD133 nanoparticles to effect separation. The labeling with and removal of unbound MAb reagent is performed manually. The CD34 reagent-treated cells are then processed on the device, where they are retained on a column located in a high-gradient magnetic field. Non-labeled cells flow through the column and are collected in the negative fraction. The labeled cells are recovered from the column after several automated separations and washing cycles by removing the magnetic field. The nanoparticles remain on the CD34/133-positive cells; however, they are biocompatible and may be infused into the recipient. Recently the company has come up with REAlease nanoparticles that can be removed from the targeted cells, however, these are not yet clinically approved. The CliniMACS device normally achieves purities over 90% with yields of approximately 60%. This results in the passive depletion of 4 to 6 logs of allogeneic T cells or autologous tumor cells. The device may be used with a variety of MAb directed against antigens expressed by various types (T, B, and NK cells), potentially allowing it to be used as a platform for multiple types of graft manipulation.

Miltenyi has also introduced the Prodigy device ( Fig. 103.1B ), which can automatically perform many of the steps requiring manual intervention on the CliniMACS. Additional features provide the potential ability to fully automate the preparation of a variety of cellular therapy products (e.g., CAR-T cells) in a functionally closed system. The regulatory status of these devices should be discussed with the appropriate regulatory agencies before clinical use.

Positive selection techniques may, however, passively deplete from the graft certain cells that could be of potential benefit to the recipient. These include some stromal elements, GVT-mediating T cells, and other populations that may facilitate engraftment. As our understanding of the identity of these populations improves, it may be possible to recover them from the normally discarded negative fraction and add them back to the CD34 + cells or to administer them in the post-transplant period as donor leukocyte infusions).

Table 103.3 lists various methods for manipulation of HSC grafts to achieve tumor removal from autologous products and T cells depletion by HPC enrichment.

Evaluation of Manipulated Allogeneic Grafts

Most allograft engineering has focused on T-lymphocyte depletion and has emphasized quantitative versus qualitative removal. The majority of allografts are infused immediately after preparation rather than after cryopreservation and storage. This restricts the types of assays that can be used to evaluate graft composition to those that have a fast turnaround since the implications of infusing large numbers of T lymphocytes can be severe or lethal. Therefore, it is important to have available methods that can rapidly enumerate the numbers of T lymphocytes within the graft. Early methods used detection of E-rosette–forming cells or manual immunofluorescence after staining with pan-T-lymphocyte–directed monoclonal antibody, however, most laboratories now rely on flow cytometry. This technology is widely used in routine clinical laboratories; however, some precautions must be taken when it is used for T-depleted allografts.

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