Chimeric antigen receptor T-cells in New Zealand: challenges and opportunities

Page created by Daniel Wang
 
CONTINUE READING
Chimeric antigen receptor T-cells in New Zealand: challenges and opportunities
viewpoint

           Chimeric antigen
          receptor T-cells in
        New Zealand: challenges
          and opportunities
 Robert Weinkove, Philip George, Myra Ruka, Tia Huia Haira, Giulia Giunti

                                              ABSTRACT
Chimeric antigen receptor (CAR) T-cells are a personalised cell and gene therapy for cancer that are becoming
an international standard of care for some refractory B-cell leukaemias, non-Hodgkin lymphomas and
myeloma. A single CAR T-cell administration can result in durable complete response for some recipients.
Domestic CAR T-cell manufacturing capability was established for Aotearoa New Zealand’s first CAR T-cell
trial (ENABLE, ClinicalTrials.gov NCT04049513). This article outlines CAR T-cell manufacturing and logistical
considerations, with a focus on New Zealand’s environment for this personalised cell and gene therapy.
We discuss Māori engagement in CAR T-cell trial and clinical service design, and propose enhancing Māori
guardianship (kaitiakitanga) over cells and genetic material through on-shore manufacture. Strategies
to safely deliver CAR T-cells within New Zealand’s healthcare system are outlined. Finally, we discuss
challenges to, and opportunities for, widening CAR T-cell availability and assuring equity of access. Based on
our experience, we consider Aotearoa New Zealand to be in an excellent position to develop and implement
investigational and commercial CAR T-cell therapies in the future.

C
       himeric antigen receptor (CAR) T-cells             As a personalised cell and gene therapy,
       are an emerging modality of can-                 CAR T-cells present unique regulatory
       cer therapy. CAR T-cells are patient             requirements. The logistics of CAR T-cell
lymphocytes that have been engineered to                production and delivery, and the potential
express a synthetic receptor, which enables             for specific adverse events, demand prepa-
them to target tumour cells.1 Internation-              ration by treatment sites. The nature of CAR
ally, CAR T-cell therapies are becoming a               T-cell therapies may raise ethical and equity
standard of care. They are a potentially                issues that need to be addressed to ensure
curative treatment for certain relapsed                 equity of access and outcomes.
and refractory (r/r) B-cell malignancies. In               We recently established local Good Manu-
adult patients with r/r B-cell lymphomas                facturing Practice (GMP) manufacture
and leukaemias, overall response rates of               of lentiviral vectors and CAR T-cells and
73–83 % and complete response rates of                  commenced enrolment to New Zealand’s
51–83 % have been reported following CAR                first CAR T-cell trial, ENABLE (Clinical-
T-cell therapy.2–4 These responses can be               Trials.gov reference NCT04049513).6 This
durable, with 75% of patients with agres-               involved close liaison with regulators and
sive B-cell lymphoma who respond at three               stakeholders and multidisciplinary input
months reporting being progression free                 for safe clinical delivery of CAR T-cell ther-
at 24 months following CAR T-cell therapy.3             apies. Here we build on this experience
Promising results have also been reported               by outlining the regulatory landscape
for other malignancies, notably myeloma.5               for CAR T-cell therapies in New Zealand,
CAR T-cell therapies involve a single cell              summarising the preparation of sites and
administration, offer curative potential and            discussing the opportunities to broaden
can often be delivered in a near-hospital               access to this new cancer treatment
outpatient setting.                                     modality in New Zealand.

                                                   96
                                                                                         NZMJ 17 September 2021, Vol 134 No 1542
                                                                                         ISSN 1175-8716      © NZMA
                                                                                         www.nzma.org.nz/journal
viewpoint

         CAR T-cell                             or natural disaster. Māori regard tissue
                                                and genetic material as a taonga (precious
       manufacturing                            item), and international manufacture limits

        and logistics                           opportunities for Māori engagement and
                                                kaitiakitanga (guardianship) over the cells
Leukocyte harvest and                           and the genetic material they contain11—
leukapheresis                                   the destination(s) and use of the cells must
  Figure 1 illustrates a typical CAR T-cell     be made clear during consent processes.
manufacturing process. In brief, white          In our view, future CAR T-cell manu-
blood cells are collected using an automated    facture within Aotearoa, either of locally
apheresis machine or by venesection of          developed products or through distributed
whole blood. Following initial processing       manufacture of commercial products,12
and purification of T-cells, a new gene (the    would improve opportunities for Māori
“transgene”) encoding the CAR is introduced,    engagement in the governance of tissue use
typically using a lentiviral or retroviral      and disposal.11
vector.7 CAR T-cells are expanded and then      Manufacture and release of
either administered fresh or cryopreserved
                                                CAR T-cells
following quality control testing.8
                                                  Although not viable except under
   Commercially licensed autologous CAR         specialised laboratory culture conditions or
T-cell products, such as tisagenlecleucel       within the intended recipient (ie, the patient
(tisa-cel) and axicabtagene ciloleucel (axi-    who underwent white blood cell collection
cel), require a leukapheresis procedure.7       used for the manufacture of that specific
The New Zealand Blood Service (NZBS) is         CAR T-cell product), CAR T-cells are defined
a national service that routinely performs      as genetically-modified organisms (GMOs)
leukapheresis for haematopoietic stem cell      by New Zealand legislation. Therefore,
harvest across New Zealand. The NZBS is         CAR T-cell manufacture is regulated by the
licensed by Medsafe to collect and manu-        Environmental Protection Authority (EPA)
facture therapeutic cells by apheresis and      under the Hazardous Substances and New
has conducted leukapheresis procedures for      Organisms (HSNO) Act 1996 (Table 1).
cellular therapy trials, including our own
                                                  Before granting approval to manufacture
ENABLE CAR T-cell trial.9,6,10 Leukapheresis
                                                CAR T-cells for the ENABLE trial, the EPA
product testing for blood-borne viruses is
                                                sought assurance that the cells could not
routinely performed at the Medsafe-licensed
                                                escape into the environment. To meet these
NZBS Donation Accreditation Laboratory.
                                                containment requirements, we modified our
For the ENABLE trial, the leukapheresis
                                                GMP manufacturing facility and its oper-
procedure and leukapheresis product testing
                                                ating procedures. The facility is inspected
is funded from the trial research budget.
                                                and audited by the Ministry for Primary
Manufacturers of commercial and inves-
                                                Industries (MPI) and Medsafe.
tigational CAR T-cell products are likely
to have specific requirements for patient          CAR T-cells are manufactured according
identification, infectious agent testing,       to validated protocols and must meet
leukapheresis performance and product           the requirements of the Pharmaceutical
labelling and shipping—and in our opinion,      Inspection Convention/Pharmaceutical
the NZBS network is likely to be well placed    Inspection Co-operation Scheme (PICS)
to conduct leukapheresis or venesection         Guide to Good Manufacturing Practice
procedures for CAR T-cell manufacture in        (Table 1). Once a CAR T-cell product has
New Zealand in the future.                      been manufactured and met pre-spec-
                                                ified “release criteria,” the CAR T-cells are
  Current commercial CAR T-cell therapies
                                                retrieved from storage and prepared for
are manufactured in a limited number of
                                                delivery to the hospital.
centres globally, none of which are in New
Zealand. This offers advantages of scale,       Shipping, product traceability,
standardisation and cost, but the need for      cell storage and disposal
bidirectional international shipping of cells     Release of CAR T-cell products to the
can present a logistical challenge, espe-       hospital site for administration to patients
cially if travel is disrupted by pandemic       also required EPA approval. Measures to

                                            97
                                                                             NZMJ 17 September 2021, Vol 134 No 1542
                                                                             ISSN 1175-8716      © NZMA
                                                                             www.nzma.org.nz/journal
viewpoint

Figure 1: Outline of CAR T-cell manufacture. Manufacture of CAR T-cells involves white blood cell harvest, typically by leukapheresis (1), followed by purification of T-cells,
and transduction of the T-cells (2), typically using a lentiviral or retroviral vector. Transduced cells express a CAR directed against a tumour antigen (3), and are expanded in
vitro in the presence of cytokines (4). Once CAR T-cell product release testing is complete, the patient received lymphodepleting (conditioning) chemotherapy (5), followed by
intravenous administration of CAR T-cells (6). The recipient is then monitored for toxicities.

                                                                                     98
                                                                                                                                                    NZMJ 17 September 2021, Vol 134 No 1542
                                                                                                                                                    ISSN 1175-8716      © NZMA
                                                                                                                                                    www.nzma.org.nz/journal
viewpoint

Table 1: Regulatory requirements for manufacture, release and delivery of CAR T-cell therapy in New Zealand.

 Regulatory body      Application to CAR T-cell therapy    New Zealand legislation or guidance                        Standards to be met

 Health and           Approval required before conduct-    Medicines Act 1981, the Guideline on the Regulation of     Conduct according to CHMP† guidance document EMA/CHMP/
 Disability Ethics    ing CAR T-cell clinical trials.      Therapeutic Products in New Zealand*                       IHC/135/95 Guideline for Good Clinical Practice E6(R2) to meet Interna-
 Committee                                                 Standard Operating Procedures for Health and Disabil-      tional Conference on Harmonisation Good Clinical Practice (IHC GCP)
 (HDEC)                                                    ity Ethics Committees, v 2.0, August 2014                  criteria.

 Gene Technology      As a cell therapy comprising         Section 30 of the Medicines Act 1981                       Demonstration of:
 Advisory             gene-modified cells, CAR T-cells                                                                •  clinical benefit (or scientific rationale for potential benefit, if
 Committee            require GTAC approval.                                                                             investigational)
 (GTAC) ‡                                                                                                             •  acceptable safety and toxicity data
                                                                                                                      •  risk assessments and risk mitigation procedures in place
                                                                                                                      •  if investigational, qualifications and experience of investigators
                                                                                                                         suitable.

 Māori                Consent and equity of access.        Te Ara Tika Guidelines for Māori research ethics, Health   Equity of access to therapy, including for those living distant from treat-
 consultation                                              Research Council New Zealand                               ment centres.
                      Māori consultation is an ethical
                      and legislative requirement for      Ministry of Health document- Equity of Healthcare for      Consent to treatment, including consent to cell shipment and storage,
                      research carried out within New      Māori: A framework                                         and future use of tissue.
                      Zealand’s district health boards .
                                                           The Treaty of Waitangi                                     Māori consultation processes for research vary regionally.

                                                           Guidance about consent with respect to the Human
                                                           Tissue Act

                                                                                           99
                                                                                                                                                            NZMJ 17 September 2021, Vol 134 No 1542
                                                                                                                                                            ISSN 1175-8716      © NZMA
                                                                                                                                                            www.nzma.org.nz/journal
viewpoint

Table 1: Regulatory requirements for manufacture, release and delivery of CAR T-cell therapy in New Zealand (continued).

 Regulatory body           Application to CAR T-cell therapy           New Zealand legislation or guidance                   Standards to be met

 Environmental             Approval required to:                       Hazardous Substances and New Organisms (HZNO)         •   Demonstrate satisfactory containment level in place to prevent
 Protection                •   manufacture CAR T-cells,                Act 1996, Section 40                                      escape of the GMOs into the environment.
 Authority (EPA)                classified as GMOs (geneti-                                                                  •   Demonstrate negligible risk of GMO forming a self-sustaining popu-
                                cally modified organisms) in                                                                     lation outside of containment when released.
                                containment                                                                                  •   Ensure necessary controls to mitigate potential risk are in place to
                           •    release CAR T-cells from                                                                         release GMO from containment.
                                containment to treatment
                                delivery site and to clinical
                                laboratory for safety testing.

 Medsafe                   License to manufacture cell thera-          PIC/S§                                                Ensuring the manufacturing facility and manufacturing procedures (in-
                           py product (pack, label and sell by                                                               cluding batch manufacturing records and product release criteria) meet
                                                                       Guide for Good Manufacturing Practice for Medicinal
                           wholesale).                                                                                       Good Manufacturing Practice (GMP) standards.
                                                                       Products, annexes 13 and 14
                           Licenses the New Zealand Blood                                                                    Leukapheresis service audited against the code of GMP.
                           Service to collect and manufacture
                           therapeutic cells by apheresis.

  *Under this legislation all clinical trials in New Zealand must receive HDEC approval.
  †CHMP (The Committee for Medicinal Products for Human Use) is the European Medicines Agency’s (EMA)
  committee responsible for human medicines.
  ‡ A committee maintained by the Health Research Council (HRC) of New Zealand to consider applications
  for trials involving gene or other biotechnology therapies.
  § PIC/S Pharmaceutical Inspection Convention Pharmaceutical Inspection Co-operation Scheme.

                                                                                                    100
                                                                                                                                                                 NZMJ 17 September 2021, Vol 134 No 1542
                                                                                                                                                                 ISSN 1175-8716      © NZMA
                                                                                                                                                                 www.nzma.org.nz/journal
viewpoint

assure safe release within the ENABLE           health outcomes, and Māori consultation is
trial included rigorous product labelling,      an ethical and legislative requirement16,17 for
shipping and traceability measures. The         research carried out within district health
International Society of Blood Transfusion      boards. Yet significant health inequities
(ISBT) standards recommend standardised         exist. Māori are approximately 20% more
bar codes to allow blood products to be         likely to develop cancer than non-Māori
shipped internationally with clear, unam-       and are twice as likely to die from cancer,14
biguous labelling and help to overcome          including from non-Hodgkin lymphoma.15
language barriers.13 Furthermore, there         These inequities are driven by delays in
are stringent procedures for packaging,         diagnosis and treatment, worse access to the
transport, administration and disposal.         wider determinants of health and inequities
   Rigorous identity checks using inde-         embedded in health system design.
pendent patient and product identifiers           Cancer research has the opportunity to
are performed and recorded on designated        contribute to Māori health advancement.
forms, to ensure the “chain of custody”         The Health Research Council (HRC) of
is maintained and that the right patient        New Zealand has implemented the Māori
receives the right product. Unused CAR          Health Advancement Guidelines, which
T-cell vial(s) are returned to the manufac-     require meaningful collaboration, consul-
turing facility for safe disposal.              tation and partnership with Māori at the
  The number and expansion of T-cells           research design stage.16 Māori expertise
varies widely between patients, so the          within the research team is recommended to
manufacturing facility often has an excess      ensure the research results in Māori health
of lymphocytes and/or CAR T-cells following     advancement and that tikanga Māori (Māori
product manufacture. Some cells must be         processes and protocol) is valued in the
retained for quality control purposes, and      research design and the sharing of results.
unless made available for future research       This framework provides an excellent
(which requires proper consent), the            resource for all health research conducted
remaining cells are typically destroyed.        in Aotearoa New Zealand.
For our own CAR T-cell trial (ENABLE), we         If access were equitable, investigational
seek consent to store unused cells within an    CAR-T cell therapies within Aotearoa New
Immune Tissue Bank. This tissue bank was        Zealand would provide an opportunity
formed with input from Māori researchers,       for Māori to benefit from treatments that
and its governance group includes Māori         are not publicly funded and that would
representation. At present, unused CAR          otherwise be cost prohibitive. Māori, who
T-cells cannot be returned to their recipient   regard genetic material as tapu (sacred/
or to the recipient’s whānau due to the         restricted), may be concerned with
cell’s legal status as a GMO, although it is    the application of genetic engineering.
our opinion that this might be possible in a    However, where there are direct health
domestic CAR T-cell manufacturing facility      benefits for Māori, and provided that these
in future, with appropriate risk assessment     benefits are well communicated, many
and controls.                                   will be supportive of this application.11,17 A
  Table 1 summarises the agencies involved      Māori consultation process is essential for
in regulating CAR T-cell manufature, release    CAR T-cell research and standard of care
and delivery in New Zealand.                    delivery, and this should include devel-
                                                opment of educational materials for Māori,
CAR-T cell therapies and equity                 their whānau and their clinicians.
for Māori                                         For our own trial (ENABLE), Māori consul-
  In Aotearoa New Zealand, Māori are            tation was sought at an early stage during
afforded the right to equitable health          protocol development. In order to address
outcomes under Te Tiriti o Waitangi and         issues relating to Māori involvement and
ethical obligations under the United Nations    recruitment and contribute to Māori health
Declaration on the Rights of Indigenous         development, our CAR T-cell researchers
Peoples. Clinicians in New Zealand have an      met with Research Advisory Group Māori
obligation to consider the degree to which      (RAG-M) at Capital and Coast District Health
they can contribute to improving Māori          Board (CCDHB) and Te Urungi Māori, the

                                           101
                                                                             NZMJ 17 September 2021, Vol 134 No 1542
                                                                             ISSN 1175-8716      © NZMA
                                                                             www.nzma.org.nz/journal
viewpoint

Māori steering group at the Malaghan            The CARTOX group was also consulted
Institute. Following consultation, a brief      regarding CAR T-cell toxicity education and
patient-focussed summary, in both English       training material for clinical staff. Since
and te reo Māori, and a visual represen-        commencing the ENABLE trial, the CARTOX
tation of the CAR T-cell treatment process      group have met at least every six months in
(Figure 1) were incorporated into the           order to review CAR T-cell-related toxicities
participant information and consent form.       reported in the ENABLE trial and to evaluate
ENABLE participants are able to donate          up-to-date published guidance on CAR T-cell
tissue to an Immune Tissue Bank, and we         toxicity management. This has led to modifi-
have consulted with our (Māori) Tissue          cations of both CRS and ICANS management
Bank Manager at the Malaghan Institute          pathways as well the production of a
regarding tissue retention and karakia          working document reviewing the evidence
arrangements. We collaborated with a            for second-line management of severe CAR
Māori clinician and researcher familiar         T-cell-related toxicities. Given our early
with managing patients with haemato-            experience and feedback from interna-
logical malignancies. To reduce the risk that   tional CAR T-cell treating clinicians and , we
distance from the treatment centre impedes      recommend implementation of a CARTOX
study access, an agreement was reached          group (or similar) for other centres adminis-
with a national cancer charity to support       tering CAR T-cell therapies.
the travel costs for study participants and a
                                                CAR T-cell toxicity education
family member, if they were not eligible for
                                                  To safely deliver CAR T-cell therapy,
the National Travel Assistance Scheme.
                                                clinical teams must be trained in the
                                                recognition and management of CAR
   Clinical delivery of                         T-cell-related toxicities. For commercial
   CAR T-cell therapy                           products, specific risk evaluation and miti-
                                                gation strategies (REMS) may be required
CAR T-cell toxicity working group               by the manufacturer, which are likely
  CAR T-cell therapies can cause specific       to involve product-specific training and
toxicities, particularly cytokine release       knowledge assessments for specific clinical
syndrome (CRS) and immune effector              staff.21 Training typically encompasses
cell-associated neurotoxicity syndrome          areas such as prescribing, dispensing and
(ICANS; neurotoxicity).18 The consensus         administering CAR T-cells and monitoring
guidelines for the diagnosis and grading of     for, recognising and treating CAR T-cell
CRS and ICANS published by the American         related toxicities.21 For the ENABLE Trial,
Society for Transplantation and Cellular        we developed an educational tool and
Therapy (ASTCT) can be readily applied in       competency assessment for clinical staff in
New Zealand hospitals.19 Consensus recom-       our inpatient and day-ward areas and for
mendations for the management of CRS and        members of the Patient at Risk and intensive
ICANS are also available.18,20                  care teams. Regular educational updates are
   For the ENABLE trial, we convened a CAR      held. One-page CRS and ICANS “quicksheets”
T-cell toxicity (CARTOX) working group,         were produced, and are displayed promi-
which included representatives from             nently in the clinical area in which a CAR
haematology, neurology, intensive care,         T-cell recipient is being treated.
immunology and nursing. The CARTOX
                                                Availability of anti-cytokine therapy
group developed and reviewed CRS and
                                                   The hallmark of CRS is fever caused by
ICANS recognition and management
                                                high levels of interleukin-6 (IL-6) during
pathways, based on consensus recommen-
                                                CAR T-cell recognition of, and activation by,
dations18 and advice from experts in the
                                                tumour cells. Tocilizumab, a monoclonal
field (Neelapu and Turtle, personal commu-
                                                antibody against the IL-6 receptor, is highly
nications). These management pathways
                                                effective for management of CRS and can be
have been posted on the hospital intranet
                                                useful for ICANS.20 Although tocilizumab is
at the CAR T-cell treating site, with clear
                                                not licensed by Medsafe in New Zealand for
paper-based management algorithms being
                                                the treatment CAR T-cell therapy toxicities,
available for easy reference on the haema-
                                                it is registered by the US Food and Drugs
tology ward where patients are treated.
                                                Administration (FDA) for this indication22

                                           102
                                                                            NZMJ 17 September 2021, Vol 134 No 1542
                                                                            ISSN 1175-8716      © NZMA
                                                                            www.nzma.org.nz/journal
viewpoint

and is universally recommended in interna-          T-cell treatment centre. After 3–4 weeks,
tional guidelines for CRS and ICANS.18,20 The       most CAR T-cell recipients will be able to
REMS strategy for axicabtagene ciloleucel           return to a home further afield. Recipient
requires that at least two doses of tocili-         education is key. Because their primary and
zumab are available on site for each patient        secondary care clinicians may lack CAR
when they are treated.21 Tocilizumab was            T-cell experience, recipients should also
alraedy approved for treating paediatric            receive clear and accurate information to
Acute Lymphoblastic Leukaemia (ALL) trial           pass on to other healthcare professionals
participants, so a clinician-led application to     they may meet.28 For the ENABLE Trial, CAR
PHARMAC was made in order to extend this            T-cell recipients are given a wallet emer-
existing approval to include ENABLE trial           gency card and a discharge summary sheet
participants. This application was successful,      (Supplementary Materials).
and PHARMAC will now fund up to three
                                                    Cellular therapy registries
doses of tocilizumab to treat CRS and/or
                                                       As CAR T-cells are a relatively new
ICANS in ENABLE trial participants.23 In our
                                                    treatment modality, there is a possibility
opinion, emergency availability of anti-IL-6
                                                    that low incidence or late onset toxicities
therapies will need to be assured for future
                                                    will emerge, such as second malignancies
investigational or commercial CAR T-cell
                                                    or adverse pregnancy outcomes. Cellular
therapies in New Zealand. High-dose cortico-
                                                    therapy registries provide an important
steroids are used for severe CRS and ICANS;
                                                    framework for long-term follow-up and
there are no restrictions on the use of these
                                                    detection of such risks.29 The Center for
in New Zealand.
                                                    International Blood and Marrow Transplant
   Expert opinion, preclinical data and             Research (CIBMTR), to which New Zealand
case reports have recommended anakinra,             stem cell transplant centres already report
siltuximab and dasatanib as second-line             safety and efficacy data following stem cell
treatments for severe CRS or ICANS.24–26            transplantation, opened its Cellular Ther-
Although there are no definitive recom-             apies Registry in June 2016, which aims to
mendations on which second-line agent to            standardise CAR T-cell toxicity reporting
use first, many sites use anakinra (a human         and data collection.29 Participation in the
intereukin-1 receptor antagonist) in this           CIBMTR Cellular Therapies Registry is open
setting because pre-clinical studies have           to CAR T-cell treatment centres worldwide,
suggested that IL-1 plays a crucial role in         provided ethical approval for data collection
the pathogenesis of CRS and neurotoxicity,          and sharing is in place. Within Australasia,
and because of anakinra’s clinical efficacy         the Australian Bone Marrow Transplant
in similar settings, such as for macrophage         Recipient Registry (ABMTRR) has recorded
activation syndrome (MAS), which can have           outcomes of haematopoietic stem cell trans-
features that overlap with severe CRS.26,27         plantation since 1992 and has expanded
In conjunction with the CARTOX group, we            its remit to collect data from CAR T-cell
developed contingency plans for second-line         recipients.30
treatment of severe CRS or ICANS, which
included the study sponsor purchasing
a supply of anakinra to be stored at the
                                                           Challenges and
treatment site.                                             opportunities
Patient education                                     In many respects, New Zealand’s
   Patients and their whānau need to receive        healthcare system is well-suited to the
education about the potential adverse               delivery of CAR T-cell therapies for haema-
effects of CAR T-cell therapy. As CRS and           tological malignancies. Although our
ICANS are typically early toxicities, it is often   regulatory processes are unique, they
recommended that patients remain close              have been successfully tested for CAR
to the CAR T-cell treating centre, alongside        T-cells through the ENABLE trial. The
a support person, for at least 21 days after        New Zealand Blood Service (NZBS) runs a
CAR T-cell therapy.28 We provide in-person          national network of blood collection and
and written education on CAR T-cell toxic-          leukapheresis centres, and NZBS electronic
ities for recipients and their support person,      systems and National Health Index (NHI)
as well as 24/7 contact details for the CAR         numbers facilitate vein-to-vein traceability

                                             103
                                                                               NZMJ 17 September 2021, Vol 134 No 1542
                                                                               ISSN 1175-8716      © NZMA
                                                                               www.nzma.org.nz/journal
viewpoint

of transfused products, follow-up and the                   needed to address this are in place, and
recording of national healthcare alerts.                    some require strengthening. A network of
Clinical haematology services benefit from                  sites offering CAR T-cell therapies across
a national network of bone marrow trans-                    Aotearoa New Zealand will reduce travel
plantation centres, with established referral               requirements for patients, but the nature of
pathways, and with comprehensive on-site                    CAR T-cell therapy means that most recip-
services, including intensive care, neurology,              ients will have to spend time away from
immunology, infectious diseases, oncology                   home; travel and accommodation support
pharmacy and specialist laboratory services.                will be essential. The need to travel is offset
Although commercial CAR T-cells are manu-                   by the one-off nature of CAR T-cell delivery
factured in large global hubs at present,                   and the fact that its principal toxicities occur
the capability to manufacture CAR T-cells                   early after administration.
within New Zealand will facilitate Māori                       Cost is a barrier to commercial CAR T-cell
engagement and guardianship and could                       therapies at present, and list prices for the
offer logistical advantages for CAR T-cell                  licensed autologous CAR T-cell products
delivery.31 Furthermore, future develop-                    axicabtagene ciloleucel and tisagenlecleucel
ments, such as outpatient management                        are high, at US$373,000 and US$475,000,
following CAR T-cell treatment and the use                  respectively.33 However, increasing compe-
of automated CAR T-cell product manufac-                    tition between manufacturers, negotiations
turing systems, may be means of lowering                    including schemes that link payment to
the cost of delivering treatment in New                     clinical response and the likely future
Zealand.32                                                  availability of tocilizumab and anakinra
  Equity of access is a key consideration                   biosimilars are expected to lower total
for all new therapies. Some mechanisms                      treatment costs in future. Pending funding

Table 2: Opportunities and challenges for CAR T-cell therapies in New Zealand.

 Challenges                        Opportunities

 Unique regulatory envi-           Medsafe regulations are harmonised with EMA*; ENABLE trial provides
 ronment                           regulatory experience

 Logistics of cell harvest         National leukapheresis & blood collection network (NZBS†); national pa-
 and manufacturing                 tient identifiers (NHI‡); domestic CAR T-cell and lentiviral vector manufac-
                                   turing capability (MIMR§; small-scale)

 Equity of access                  Involve Māori in service planning; leverage national stem cell transplant
                                   network; one-off CAR T-cell therapy possibly preferable to ongoing treat-
                                   ments; inter-district funding and national travel assistance schemes

 High cost of commercial           CAR T-cells are a “one-off” rather than ongoing cost; declining costs due to
 CAR T-cell therapies              competition; clinical trial access pending commercial delivery; outpatient
                                   administration can lower costs

 Limited clinician ex-             National network of stem cell transplant centres; clinical trial experience
 perience of CAR T-cell            of CAR T-cell delivery (ENABLE)
 delivery

 Restrictions on medicines         Tocilizumab and anakinra biosimilars are in development; PHARMAC
 used to treat CAR T-cell          provides a national funding mechanism (“Special Authority”)
 toxicities

  * European Medicines Agency (EMA).
  †New Zealand Blood Service (NZBS).
  ‡ National Health Index (NHI).
  § Malaghan Institute of Medical Research (MIMR

                                                     104
                                                                                            NZMJ 17 September 2021, Vol 134 No 1542
                                                                                            ISSN 1175-8716      © NZMA
                                                                                            www.nzma.org.nz/journal
viewpoint

of CAR T-cell therapies, clinical trials can     programme in New Zealand, with particular
help bridge the gap in availability, at least    consideration to Māori engagement and
for some patients, and prepare our regu-         access to treatment. Having reviewed the
latory and clinical systems for routine CAR      challenges and opportunities, we believe
T-cell delivery. Table 2 summarises key          Aotearoa New Zealand is in an excellent
challenges and opportunities for CAR T-cell      position to develop and implement both
therapies in New Zealand.                        investigational and commercial CAR T-cell
                                                 therapies in the future.
          Conclusions
  CAR T-cell therapy is shifting the treatment         Supplementary
paradigm of relapsed and/or refractory
B-cell malignancies internationally. In this
                                                          material
                                                  •   Supplementary Materia 1: CAR T-cell
viewpoint, we have outlined the regulatory
                                                      Wallet Emergency Card
processes and clinical preparations involved
in establishing the first CAR T-cell therapy      •   Supplementary Material 2: ENABLE
                                                      Trial Discharge Summary Sheet

                                           105
                                                                          NZMJ 17 September 2021, Vol 134 No 1542
                                                                          ISSN 1175-8716      © NZMA
                                                                          www.nzma.org.nz/journal
viewpoint

                                         Competing interests:
         RW, PG, TH and GG are employees of the Malaghan Institute of Medical Research,
        which sponsors the ENABLE trial. The authors do not have proprietary or financial
                         interests in the WZTL-002 CAR T-cell product.
                                          Acknowledgements:
             The authors acknowledge funding support from the Health Research
             Council of New Zealand (19/139 and 19/816), the Ministry of Business
        Innovations and Employment, the LifeBlood Trust and Freemasons New Zealand.
                 This manuscript is dedicated to the memory of Dr John Carter.
                                          Author information:
       Robert Weinkove: Cancer Immunotherapy Programme, Malaghan Institute of
 Medical Research, Wellington, New Zealand; Wellington Blood & Cancer Centre, Capital &
Coast District Health Board, Wellington, New Zealand; Department of Pathology & Molecular
             Medicine, University of Otago Wellington, Wellington, New Zealand.
     Philip George: Cancer Immunotherapy Programme, Malaghan Institute of Medical
      Research, Wellington, New Zealand; Wellington Blood & Cancer Centre, Capital &
                    Coast District Health Board, Wellington, New Zealand.
      Myra Ruka: Haematology Department, Waikato District Health Board, Hamilton,
     New Zealand; Hei Āhuru Mōwai – Māori Cancer Leadership Aotearoa, Wellington,
    New Zealand; Te Aho o Te Kahu – Cancer Control Agency, Wellington, New Zealand.
         Tia Huia Haira: Cancer Immunotherapy Programme, Malaghan Institute of
   Medical Research, Wellington, New Zealand; Clinical Human Immunology Laboratory,
              Malaghan Institute of Medical Research, Wellington, New Zealand.
     Giulia Giunti: Cancer Immunotherapy Programme, Malaghan Institute of Medical
       Research, Wellington, New Zealand; Clinical Human Immunology Laboratory,
              Malaghan Institute of Medical Research, Wellington, New Zealand.
                                         Corresponding author:
      Dr Robert Weinkove, Malaghan Institute of Medical Research, PO Box 7060, Wellington,
                   New Zealand, +64 4 499 6914 (phone), +64 4 499 6915 (fax)
                             Email: rweinkove@malaghan.org.nz
                                                   URL:
     www.nzma.org.nz/journal-articles/chimeric-antigen-receptor-t-cells-in-new-zealand-chal-
                                   lenges-and-opportunities

REFERENCES
1. Oldham RAA, Medin JA.                  Online First: 2020/09/06]           Ganzetti M, Fernández de
    Practical considerations for    3.    Locke FL, Ghobadi A,                Larrea C, et al. CAR T-Cells
    chimeric antigen receptor             Jacobson CA, et al. Long-           in Multiple Myeloma:
    design and delivery.                  term safety and activity            State of the Art and Future
    Expert Opin Biol Ther                 of axicabtagene ciloleucel          Directions. Front Oncol
    2017;17(8):961-78. doi:               in refractory large B-cell          2020;10:1243. doi: 10.3389/
    10.1080/147125                        lymphoma (ZUMA-1): a                fonc.2020.01243 [published
    98.2017.1339687 [published            single-arm, multicentre,            Online First: 2020/08/28]
    Online First: 2017/06/07]             phase 1-2 trial. Lancet        6.   George P, Dasyam N,
2.     Abramson JS, Palomba               Oncol 2019;20(1):31-42.             Giunti G, et al. Third-gen-
       ML, Gordon LI, et al.              doi: 10.1016/s1470-                 eration anti-CD19 chimeric
       Lisocabtagene mara-                2045(18)30864-7 [published          antigen receptor T-cells
       leucel for patients with           Online First: 2018/12/07]           incorporating a TLR2
       relapsed or refractory       4.    Park JH, Riviere I, Gonen M,        domain for relapsed or
       large B-cell lymphomas             et al. Long-Term Follow-            refractory B-cell lympho-
       (TRANSCEND NHL 001):               up of CD19 CAR Therapy              ma: a phase I clinical trial
       a multicentre seamless             in Acute Lymphoblastic              protocol (ENABLE). BMJ
       design study. Lancet               Leukemia. N Engl J Med              Open 2020;10(2):e034629.
       2020;396(10254):839-52.            2018;378(5):449-59. doi:            doi: 10.1136/bmjop-
       doi: 10.1016/s0140-                10.1056/NEJMoa1709919               en-2019-034629 [published
       6736(20)31366-0 [published                                             Online First: 2020/02/12]
                                    5.    Rodríguez-Lobato LG,

                                                 106
                                                                                      NZMJ 17 September 2021, Vol 134 No 1542
                                                                                      ISSN 1175-8716      © NZMA
                                                                                      www.nzma.org.nz/journal
viewpoint

7.   Levine BL, Miskin J,          14. Teng AM, Atkinson J,              [published Online
     Wonnacott K, et al.               Disney G, et al. Ethnic           First: 2018/11/09]
     Global Manufacturing              inequalities in cancer        21. Administration USFaD.
     of CAR T Cell Therapy.            incidence and mortality:          Approved risk evaluation
     Mol Ther Methods Clin             census-linked cohort stud-        and mitigation strategies
     Dev 2017;4:92-101. doi:           ies with 87 million years         (REMS). Yescarta (axicabta-
     10.1016/j.omtm.2016.12.006        of person-time follow-up.         gene ciloleucel), 2018.
     [published Online                 BMC Cancer 2016;16(1):755.
                                                                     22. Le RQ, Li L, Yuan W, et al.
     First: 2017/03/28]                doi: 10.1186/s12885-016-
                                                                         FDA Approval Summary:
8.   Roddie C, O’Reilly M,             2781-4 [published Online
                                                                         Tocilizumab for Treatment
     Dias Alves Pinto J, et al.        First: 2016/09/28]
                                                                         of Chimeric Antigen
     Manufacturing chimeric        15. Gurney J, Stanley J, McLeod       Receptor T Cell-Induced
     antigen receptor T cells:         M, et al. Disparities in          Severe or Life-Threat-
     issues and challenges.            Cancer-Specific Survival          ening Cytokine Release
     Cytotherapy 2019;21(3):327-       Between Māori and                 Syndrome. Oncologist 2018
     40. doi: 10.1016/j.               Non-Māori New Zealand-            doi: 10.1634/theoncolo-
     jcyt.2018.11.009 [published       ers, 2007-2016. JCO Glob          gist.2018-0028 [published
     Online First: 2019/01/28]         Oncol 2020;6:766-74.              Online First: 2018/04/07]
9.   Health Mo. Medsafe                doi: 10.1200/go.20.00028
                                                                     23. Agency PM. New Zealand
     Licensed Medicine                 [published Online
                                                                         Pharmaceutical Schedule.
     Manufacturing Sites 2020.         First: 2020/06/09]
                                                                         In: PHARMAC, ed.: New
10. Gasser O, Sharples KJ,         16. Zealand HRCoN. Māori              Zealand Government, 2019.
    Barrow C, et al. A phase           Health Advancement
                                                                     24. Mestermann K, Giavridis T,
    I vaccination study with           Guidelines. In: Coun-
                                                                         Weber J, et al. The tyrosine
    dendritic cells loaded             cil HR, ed., 2019.
                                                                         kinase inhibitor dasatinib
    with NY-ESO-1 and              17. Lipworth W, Axler R.              acts as a pharmacologic on/
    alpha-galactosylceramide:          Towards a bioethics               off switch for CAR T cells.
    induction of polyfunctional        of innovation. J Med              Sci Transl Med 2019;11(499)
    T cells in high-risk mela-         Ethics 2016;42(7):445-9.          doi: 10.1126/scitranslmed.
    noma patients. Cancer              doi: 10.1136/medeth-              aau5907 [published
    Immunol Immunother                 ics-2015-103048 [published        Online First: 2019/07/05]
    2018;67(2):285-98. doi:            Online First: 2016/03/27]
                                                                     25. Turtle CJ, Hay KA, Hanafi
    10.1007/s00262-017-            18. Neelapu SS, Tummala S,            LA, et al. Durable Molecu-
    2085-9 [published Online           Kebriaei P, et al. Chimeric       lar Remissions in Chronic
    First: 2017/11/03]                 antigen receptor T-cell           Lymphocytic Leukemia
11. Beaton A, Smith, B., Toki,         therapy - assessment and          Treated With CD19-Specific
    V., Southey, K., & Hudson,         management of toxici-             Chimeric Antigen Recep-
    M. Engaging Māori in               ties. Nat Rev Clin Oncol          tor-Modified T Cells After
    Biobanking and Genetic             2018;15(1):47-62. doi:            Failure of Ibrutinib. J Clin
    Research: Legal, Ethical           10.1038/nrclinonc.2017.148        Oncol 2017;35(26):3010-
    and Policy Challenges.         19. Lee DW, Santomasso BD,            20. doi: 10.1200/
    The International Indige-          Locke FL, et al. ASTCT            jco.2017.72.8519 [published
    nous Policy Journal. The           Consensus Grading for             Online First: 2017/07/18]
    International Indigenous           Cytokine Release Syndrome     26. Norelli M, Camisa B,
    Policy Journal 2015;6(3)           and Neurologic Toxicity           Barbiera G, et al. Mono-
12. Harrison RP, Ruck S, Rafiq         Associated with Immune            cyte-derived IL-1 and IL-6
    QA, et al. Decentralised           Effector Cells. Biol Blood        are differentially required
    manufacturing of cell and          Marrow Transplant                 for cytokine-release
    gene therapy products:             2019;25(4):625-38. doi:           syndrome and neurotox-
    Learning from other                10.1016/j.bbmt.2018.12.758        icity due to CAR T cells.
    healthcare sectors. Biotech-       [published Online                 Nat Med 2018;24(6):739-48.
    nol Adv 2018;36(2):345-57.         First: 2018/12/29]                doi: 10.1038/s41591-018-
    doi: 10.1016/j.                20. Hay KA. Cytokine release          0036-4 [published Online
    biotechadv.2017.12.013             syndrome and neurotox-            First: 2018/05/29]
    [published Online                  icity after CD19 chimeric     27. Kumar B, Aleem S, Saleh
    First: 2017/12/27]                 antigen receptor-modified         H, et al. A Personalized
13. ICCBBA. Labelling of Blood         (CAR-) T cell therapy. Br J       Diagnostic and Treatment
    Components. ISBT 128               Haematol 2018;183(3):364-         Approach for Macrophage
    Standard: ICCBBA, 2016.            74. doi: 10.1111/bjh.15644        Activation Syndrome and

                                              107
                                                                                 NZMJ 17 September 2021, Vol 134 No 1542
                                                                                 ISSN 1175-8716      © NZMA
                                                                                 www.nzma.org.nz/journal
viewpoint

    Secondary Hemophagocytic      30. Nivison-Smith I, Bardy            Economic Evaluation
    Lymphohistiocytosis in            P, Dodds AJ, et al. A             of Chimeric Antigen
    Adults. J Clin Immunol            Review of Hematopoietic           Receptor T-Cell Therapy
    2017;37(7):638-43. doi:           Cell Transplantation              by Site of Care Among
    10.1007/s10875-017-               in Australia and New              Patients With Relapsed or
    0439-x [published Online          Zealand, 2005 to 2013. Biol       Refractory Large B-Cell
    First: 2017/09/06]                Blood Marrow Transplant           Lymphoma. JAMA Netw
28. Taylor L, Rodriguez ES,           2016;22(2):284-91. doi:           Open 2020;3(4):e202072.
    Reese A, et al. Building a        10.1016/j.bbmt.2015.09.009        doi: 10.1001/jamanetworko-
    Program: Implications for         [published Online                 pen.2020.2072 [published
    Infrastructure, Nursing           First: 2015/09/20]                Online First: 2020/04/07]
    Education, and Training for   31. Bachanova V, Bishop           33. Lin JK, Muffly LS, Spinner
    CAR T-Cell Therapy. Clin J        MR, Dahi P, et al. CAR            MA, et al. Cost Effective-
    Oncol Nurs 2019;23(2):20-         T Cell Therapy During             ness of Chimeric Antigen
    26. doi: 10.1188/19.Cjon.         the COVID-19 Pandemic.            Receptor T-Cell Therapy
    S1.20-26 [published               Biol Blood Marrow                 in Multiply Relapsed or
    Online First: 2019/03/19]         Transplant 2020 doi:              Refractory Adult Large
29. Pasquini MCP, M-A.                10.1016/j.bbmt.2020.04.008        B-Cell Lymphoma. J Clin
    Real World Data on                [published Online                 Oncol 2019;37(24):2105-19.
    CAR T-Cell Recipients:            First: 2020/04/17]                doi: 10.1200/jco.18.02079
    Are We There Yet? The         32. Lyman GH, Nguyen                  [published Online
    Hematologist 2019;16(2)           A, Snyder S, et al.               First: 2019/06/04]

                                             108
                                                                                NZMJ 17 September 2021, Vol 134 No 1542
                                                                                ISSN 1175-8716      © NZMA
                                                                                www.nzma.org.nz/journal
You can also read