kelvinryq820430的个人博客分享 http://blog.sciencenet.cn/u/kelvinryq820430

博文

[转载]Management of Adults and Children receiving CAR T-cell thera

已有 251 次阅读 2024-6-11 16:10 |个人分类:CAR-T|系统分类:论文交流|文章来源:转载

ABSTRACT:

Background: Several commercial and academic autologous chimeric antigen receptor T-cell products targeting CD19 have been approved in Europe for relapsed/refractory B-cell acute lymphoblastic leukemia, high-grade B-cell lymphoma, and mantle cell lymphoma. Products for other diseases such as multiple myeloma and follicular lymphoma are likely to be approved by the European Medicines Agency in the near future.

Design: EBMT-JACIE and the European Haematology association (EHA) proposed to draft best  practice  recommendations  based  on  the  current  literature,  to   support  healthcare professionals in delivering consistent, high-quality care in this rapidly moving field

Results: Thirty-six CAR-T experts (medical; nursing; pharmacy/laboratory) assembled to draft recommendations to cover all aspects of CAR-T patient care and supply chain management, from  patient  selection  to  long-term  follow-up,  post-authorisation  safety  surveillance  and regulatory issues.

Conclusions: We provide practical, clinically relevant recommendations on the use of these high-cost,  logistically  complex  therapies  for  hematologists/oncologists,  nurses  and  other stakeholders including pharmacists and health sector administrators involved in the delivery of CAR-T in the clinic.

 

KEYWORDS:

CART-cells (CAR-T), B-cell acutelymphoblastic leukemia (B-ALL),adult relapsed/refractory (r/r) diffuse large B-cell lymphoma (DLBCL),Multiple myeloma (MM), Mantle cell lymphoma (MCL), follicular lymphoma (FL), cytokine release syndrome (CRS), Macrophage activation syndrome (MAS), immune effector cell-associated neurotoxicity syndrome (ICANS), JACIE, long-term follow-up (LTFU).

 

 

 

INTRODUCTION:

CD19   chimeric   antigen   receptor   T-cell    (CAR-T)   therapeutics   are   widely   used   for relapsed/refractory (r/r) B-cell malignancies including acutelymphoblastic leukemia (B-ALL), large B-cell lymphoma (LBCL) and mantle cell lymphoma (MCL)1-3. CAR-T are also under evaluation in multiple myeloma, acute myeloid leukemia and solid tumors4,5 .

Three CAR-T products are licensed: Tisagenlecleucel (Kymriah®, Novartis) for r/rpaediatric B-ALL and adult LBCL; Axicabtagene ciloleucel (Yescarta®, Gilead), for r/r adult LBCL and Tecartus (brexucabtagene autoleucel), for r/r adult MCL. In the US, lisocabtagene maraleucel (Liso-Cel, BMS) is approved for r/r LBCL andidecabtagenevicleucel [ide-cel, Abecma]and JNJ-45287 have been approved for r/rmultiple myeloma4. In Spain, regulators have approved academic CD19 CAR-T (ARI-0001) for r/r B-ALL8.

CAR-T   confers   a   risk    of   potentially   life-threatening    immunological   toxicities    and comprehensive training of personnel involved in CAR-T delivery, including intensive care unit (ICU) and neurology specialists, is key9.

Post-marketing pharmacovigilance over 15 years post-infusion is mandated by EMA to ensure ongoing evaluation of efficacy and safety of licensed CAR-T in the real-world setting via the EBMT registry. CIBMTR (Center for International Blood and Marrow Transplant Research) fulfils a similar role. Further, the PASS (post-authorization safety studies) initiative makes an assessment of the value of CAR-T in relation to standard-of-care treatments.

 

METHODOLOGY

In 2021, EBMT and EHA proposed an expanded revision of the 2019 EBMT-JACIE CAR-T guidelines10,11 . Thirty-six CAR-T experts (medical; nursing; pharmacy/laboratory) assembled to draft recommendations based on the current literature, to reflect current best practice in this rapidly moving field and to support healthcare professionals in delivering consistent, high- quality care. Given the absence of randomized trial evidence, a decision was made not to grade these recommendations. They therefore represent the consensus view of the authors.

The recommendations principally apply to licensed CAR-T therapies. For CAR-T clinical trials, healthcare teams should follow relevant trial protocols.

 

 

PATIENT ELIGIBILITY

Patient eligibility should be assessed by a CAR-T centre multi-disciplinary team including cellular therapy and haematology/oncology disease specialists. Medical history, performance status and CAR-T product should be considered with respect to tolerability (Table 1).

 

 

Eligibility Criteria

EBMT/EHA

recommendations

Comments

Age limit

No age limit

Decision should be based on physical condition rather   than age, although ability to   collect sufficient cells by apheresis can   be a limiting factor in

infants and small children.

Real-world CAR-T data suggests that 5.9% of treated patients with B-ALL were <3   years old and 53.5% of treated   patients with NHL were >65 years    old and that CR rates was comparable in both groups to the rest of the

population.

Performance Status

ECOG <2, Karnofsky >   60% or Lansky >60%

Although patients with ECOG>1 were treated outside   clinical trials, it was associated   with significantly decreased OS and PFS.

 

 

 

 

Life expectancy

More than 6-8 weeks

Requires careful consideration in terms of risk-benefit ratio.

High tumour burden

Risk-benefit assessment   required

High tumor burden in B-ALL and LBCL is a risk factor for   treatment

failure and greater toxicity and careful consideration of the individual risk-   benefit ratio is required.

History of malignancy

Absence of active malignancy requiring treatment other than non-melanoma skin cancer or    carcinoma in situ (e.g. cervix,    bladder, breast).

Requires careful consideration of the risk-benefit ratio.

Prior allo-HCT

Not a contraindication

Not a contraindication when off immunosuppression but in ALL may   increase risk of CAR-T associated toxicity

Prior   treatments directed towards   antigenic target  of CAR-T e.g. bispecific antibodies/ prior CAR-T

Not a contraindication, but

antigen negative escape

should be excluded at   relapse post-targeted therapy and

prior to CAR-T especially in B-cell ALL

Reduced CD19 expression may not decrease the efficacy   of anti-CD19 CAR-T in B-ALL;   however, prior treatment with blinatumomab   may    impair efficacy

A second infusion of anti-CD19 CAR T-cells maybe feasible and can induce remission in a subset of patients.

Immunosuppressive treatment

Relative contraindication

Any systemic immunosuppressive treatment may impair the efficacy of CAR-T   Intermittent topical, inhaled, or intranasal corticosteroids are

permitted.

Bacterial or fungal infections

Active infection is a contraindication

Infection should be treated and well controlled such   that the patient should   be stable prior to leukapheresis. In most cases, active infection requires

only a temporary deferral.

Viral infection

Viremia is a contra-indication.

Treatment   should be delayed in cases of positive COVID-  19 PCR.12

Active viral infection should prompt deferral of initiation of CAR-T

therapy until the infection is controlled. Some latent infections, e.g. HIV,

are a contraindication to manufacturing for several (but not all) commercial and trial CAR-T products.

When proceeding to CAR-T in cases of latent HBV, HCV or HIV infections,   prophylactic anti-viral treatment is required.

Asymptomatic patients testing positive for COVID-19 by qPCR may

proceed to CAR-T manufacture, but this is done at risk and at the

physician’s discretion. Before proceeding, feasibility   should be checked with the CAR-T   manufacturer well in advance of leukapheresis.

History of central

nervous system (CNS) involvement

Relative contraindication

Requires careful consideration of the risk-benefit ratio.

LBCL: for ZUMA-1 and Juliet, CNS   involvement was an exclusion

criterion, but in Transcend-world, controlled SCNSL was permitted on study.

MCL: on ZUMA-2, CNS involvement was an exclusion.

B-ALL: on ELIANA, active CNS   involvement was an exclusion.

Real world evidence (RWE) on CAR-T for CNS involvement in DLBCL is   emerging: suggesting that it is well tolerated and has potential efficacy.

MM: CNS involvement was an exclusion   in KarMMa study

Table 1. Patient eligibility criteria for CAR-T

 

 

 

SCREENING LABORATORY TESTS AND IMAGING

To ensure patient eligibility and fitness, the screening tests in Table 2 should be considered. This list is not exhaustive, and, in the trial setting, trial protocols should be followed.

 

Screening tests

EBMT/EHA recommendations

Comments

Disease confirmation

Diagnosis should be confirmed using appropriate tests

e.g. histology for NHL; immunophenotyping for ALL

Haematology

Evidence of adequate bone marrow reserve

Bone marrow reserve is difficult to evaluate in high burden r/r ALL and MM.

Bilirubin

<34µmol/L in trials; higher limit acceptable (<43µmol/L) with Gilbert’s syndrome

No trial data regarding patients outside of these parameters.

 

 

 

 

AST/ALT

<4x ULN a contra-indication in some trials

Attempt   to identify cause of liver   derangement e.g.  infection, drug   toxicity including antifungals, VoD,   GvHD.

Creatinine clearance

> 30 ml/min

Physicians should consider appropriate dose

reductions in cyclophosphamide and fludarabine  when creatinine clearance is below 60ml/min   and potentially an increased   interval between LD and  CAR-T return to permit clearance of Fludarabine  metabolites.

Hepatitis B

As per national guidelines

Serology/molecular testing

Hepatitis C

As per national guidelines

Serology/molecular testing

HIV

Leukapheresis for KymriahTM manufacturing will not be accepted from patients with a positive test for

active HBV, HCV, or HIV (SPC).   This is not the case for   YescartaTM

KymriahTM employs lentiviral vectors for   CAR gene transfer whereas YescartaTM uses retroviral vectors.   There is a theoretical concern regarding   lentiviral

recombination events.

COVID-19

Nasopharyngeal PCR before leukapheresis should be negative

Asymptomatic patients testing positive for COVID-

19 by qPCR may proceed to CAR-T manufacture, but this is done at risk and at the physiciansdiscretion.

Before proceeding, feasibility should be checked with

the CAR-T manufacturer well in advance of leukapheresis.

COVID-19 vaccination

Recommended

Though data is limited, patients should be vaccinated

against COVID-19, where possible, prior to admission for CAR-T.

Cardiac function

TTE to assess cardiac function and exclude   significant pericardial effusions and structural abnormalities –

LVEF <40% (via 4DEF or Simpson’s Biplane

method) is a relative contraindication.

ECG to exclude significant arrhythmias.

Cardiac biomarkers (troponin and NT-proBNP) at baseline

CMR to assess extent disease in PMBCL with cardiac involvement.

Consider cardio-oncology review for further

assessment of treatment   suitability and scope for cardiac   optimisation

CNS imaging

MRI not required except in those with a history of CNS disease or current neurological symptoms

Lumbar puncture

Lumbar puncture not required except in those with a history of CNS disease or current neurological

symptoms

Fertility

Females of childbearing potential must have a   negative serum or urine pregnancy test

Test must be repeated and confirmed negative within

8 days of the CAR-T cell infusion

Table 2. Screening tests prior to CAR-T therapy. Key. NHL: Non-Hodgkin Lymphoma; ALL: acutelymphoblastic leukemia;  ULN: upper limit of normal;  TTE: transthoracic echocardiogram; LVEF: left ventricular ejection fraction; ECG: electrocardiogram;  CMR: cardiac magnetic resonance; PMBCL: primary mediastinal B cell lymphoma; MRI: Magnetic resonance  imaging;  CNS: central nervous system;  VoD:  veno-occlusive disease; GvHD: graft versus host disease.

 

 

WORK-UP FOR LEUKAPHERESIS

Leukapheresis procurement in the European Union must comply with the Tissue and Cell Directives (2004/23/EC; 2006/17/EC; 2006/86/EC). Shipment across borders requires current viral serology and compliance with regulations in both the countries of origin and destination 13,14

.

A pre-leukapheresis checklist and suggested washout periods for pre-leukapheresis therapeutics are listed in Tables 3 and 4.

 

 

 

Prior to Apheresis

EBMT/EHA recommendations

Comments

Performance status

ECOG <2, Karnowski > 60%

At discretion of leukapheresis practitioner

Interval following

Allow sufficient time for recovery from cytotoxic

Adequate marrow recovery from prior chemotherapy

exposure to

chemotherapy/immunosuppression/ steroids (see

required

chemotherapy

Table 4 for washout periods)

Interval following      exposure to   steroids

A minimum of 3 days   prior to leukapheresis.

Optimally, 7 days to minimise   impact on leukapheresis

Physiological replacement doses of hydrocortisone permitted,   Topical and inhalational steroids also

permitted

Blood oxygen saturation

≥ 92% on room air

Hepatitis B, Hepatitis C,   HIV, Syphilis, and HTLV

To be done within 30 days of leukapheresis. Results must be available at the time of collection and

shipment. Mandatory in some   countries

In some countries, only serological testing is

Journal Pre-proofrequired; nucleic acid testing (NAT) is not necessary if all serological testing is negative

COVID-19 PCR

Not a contraindication in asymptomatic patients. Contraindication   in symptomatic patients

Apheresis physician and manufacturing facility should be informed if positive PCR

COVID-19 vaccination

Recommended

Though data is limited, patients should be vaccinated

against COVID-19, where possible, prior to admission for CAR-T.

Standard electrolytes and renal function

Required

Leukapheresis can be complicated by electrolyte imbalance and fluid shifts   during the procedure

Haemoglobin

Hemoglobin>80 g/L recommended   Hematocrit >0.24 recommended

To help establish a good interface during   leukapheresis

Absolute Lymphocyte count (ALC)

≥ 0.2x109/L recommended

Low counts indicate insufficient haematological

recovery and may predict for production failure.

Higher count required in small children Of note,

0.2x109/L CD3+ count is the minimum recommended   threshold

Platelet count

> 30x109/L recommended

Transfuse as required,   particularly for insertion of central line prior to leukapheresis.

Full Blood Count (FBC)

To be repeated at the end of apheresis procedure

Apheresis can remove more than 30% of circulating platelets

Table 3. Checklist prior to leukapheresis

 

Type of therapy

EBMT/EHA   recommendations

Comments

Allo-HCT

Patients should be off

immunosuppression and GvHD- free

A minimum of one month is recommended with the   requirement to be GvHD-free and off

immunosuppression

DLI

At   least 4 weeks

6-8 weeks maybe safer to rule out any GvHD

High-dose   chemotherapy

3-4   weeks

Recovery from cytopenias   is required

Intrathecal   therapy

One   week

Short-acting cytotoxic/anti- proliferative drugs

3 days

Recovery from cytopenias   is required

Systemic   corticosteroids

Minimum of 3 days but   ideally 7 days

ALC ≥0.2x109/L is recommended

Table 4. Washout period before leukapheresis [adapted from Kansangra et al, BBMT 201915] Key. Allo-SCT: allogeneic stem cell transplantation; GvHD: graft versus host disease; DLI: donor lymphocyte infusion; ALC Absolute Lymphocyte Count.

 

 

 

PERFORMING LEUKAPHERESIS

To be a CAR-T delivery site, accreditation with FACT-JACIE is recommended. Pharmaceutical providers and health service commissioners may have additional requirements.

CAR-T product prescription/order and non-mobilised leukapheresis 16,17  scheduling/shipping, is  co-ordinated  with  the  CAR-T  manufacturing  facility  (often  via  proprietary  web-based platforms),

Most manufacturers stipulate storage of fresh leukapheresis at 2-8 °C prior to shipping. Novartis additionally accept locally cryopreserved starting material (within 30 months).

Accredited, validated leukapheresis testing methods should be compatible with manufacturer’s requirements   and   authorizations.   An   absolute   lymphocyte   count   (ALC)   threshold   of 0.2x109/L17,18    is   generally  recommended19,20,  but   emerging  evidence   supports   CAR-T leukapheresis in paediatric and adult patients with low ALC21,22 .

Infectious  disease  markers  must  be  tested  on  peripheral  blood  (PB)  within  30  days  of leukapheresis (with results available on the day of shipment). Microbial contamination is rare and the presence of leukemic blasts is acceptable to manufacturers.

Based on the observation that T-cells suffer qualitative damage from chemotherapy, feasibility of pre-emptive  leukapheresis  in  high-risk  patients  is  currently  being  exploredbut  with significant regulatory, infrastructural, and cost implications.

 

 

BRIDGING THERAPY

‘Bridging’  therapy,   administered  in  the   4-6  weeks  between   leukapheresis  and  CAR-T admission, aims to reduce disease burden and in doing so, improve intention-to-treat and reduce CAR-T immunotoxicity23-25.

Patient-specific  bridging  recommendations  should  be  made  by  a  multi-disciplinary  team following review of response to prior therapy, overall tumour burden, and anatomical sites of disease. Bridging can be omitted if the CAR-T ‘vein-to-vein’ time is short and the disease

burden is low. Otherwise, bridging is broadly split into: (a) high-dose chemotherapy; (b) low- dose chemotherapy; (c) radiotherapy; and (d) novel agents/approaches. Radiation therapy can be an effective bridge, but the impact on circulating lymphocytes should be considered if radiation  is  commenced prior to  leukapheresis.  Further  studies  are  warranted  to  optimise bridging approaches26.

Bridging can be delivered at the CAR-T centre or at the referring center, provided there is clear communication regarding the selected bridging strategy, management of complications, and scheduling of bridging in relation to CAR-T admission, fastidiously observing recommended washout periods (Table 5).

 

 

 

 

Type of therapy

EBMT/EHA recommendations

Comments

High-dose chemotherapy

3-4 weeks

To avoid additional toxicity and prolonged cytopenias

Intrathecal therapy

1 week

To avoid additional toxicity

Short-acting cytotoxic/anti-   proliferative drugs

3 days

To avoid additional toxicity

Radiotherapy

1 weeks (2 weeks for lung)

To avoid additional toxicity

TKI

3 days

To avoid additional toxicity

Table 5. Washout period between the bridging therapy and the onset of LD conditioning [Expert opinion]. Key:

TKI: tyrosine-kinase inhibitor.

 

 

HOSPITALIZATION

Outpatient CAR-T administration can be done safely, provided clear policies, appropriate infrastructure,  well-trained  staff,  and  capacity  for  24/7  hospitalization  in  the   event  of complications is in place14. As such facilities are not available in most European centers, we recommend that patients remain hospitalized for at least 14 days following infusion (Table 6).

 

 

Period

EBMT/EHA recommendations

Comments

Day 0 to Day +14 post-infusion

Ideally 14 days hospitalization.

Consider 10 days inpatients with no post-   infusion complications.

Outpatient follow-up is possible in centres that can

provide 24/7 contact with immediate availability of

specialist inpatient care. For this arrangement, patients should be located within   30-60 minutes of the CAR-T  centre

From hospital

discharge to Day +28 post-infusion

Patients should be located within   60 minutes of the center and the continuous presence of a

caregiver who is educated to recognize the    signs and symptoms of CRS   and ICANS is required

Delayed CRS and ICANS can emerge following discharge   from hospital

Table 6. Recommendations on hospitalisation in the first 28 days after CAR-T infusion.

 

 

LYMPHODEPLETING CONDITIONING (LD)

LD acts to enhance CAR-T proliferation by modulating cytokine and immune pathways. Fludarabine and cyclophosphamide (FC) is widely used27. Fludarabine dosing is consistent between  products  and  indications  (25-30  mg/m2/day  x3  days)  whilst  cyclophosphamide schedules differ28. Bendamustine (+/- fludarabine) has been tested in CD30CAR-T for Hodgkin Lymphoma as an alternative to FC29.

LD is administered following CAR-T product release, the week prior to CAR-T infusion with a minimum of two rest days. Where CAR-T infusion is delayed by ≥4 weeks, repeat LD is recommended, with consideration given to patient fitness, blood counts and prior fludarabine exposure10.

Potential  complications  from  LD  include  pancytopenia,  immunosuppression,  infection, neurotoxicity, haemorrhagic cystitis, pericarditis and secondary malignancy. Renal or hepatic impairment should prompt appropriate dose modification. Considerations prior to LD are outlined in Table 7.

 

 

 

Criterion

EBMT/EHA

recommendations

Comments

CAR T-cell product

LD should be administered    following receipt of CAR-T product   on site

Exceptional situations may necessitate the administration of LD

following confirmation of successful CAR-T manufacture, but prior to receipt.

Clinical conditions

Active infections should be    ruled out before starting LD

Patient should be medically fit to proceed to LD

Blood oxygen saturation

92% on air

WBC

Administer LD to all patients irrespective of WBC or ALC

The SPC for KymriahTM state that patients with low WBC (<1x109/L) 1 week prior to CAR-T   infusion may not require LD. Some investigators  use LD with caution when unexplained   neutropenia pre-dates CAR-T      admission. However, LD is important to CAR-T activity and

proceeding with CAR-T without LD is not generally recommended.

C-reactive protein, ferritin,   lactate dehydrogenase,

metabolic profiling,

fibrinogen level

Required to rule out ongoing infection.

Baseline assessments of risk for CRS and NT

Bilirubin

<34µmol/L; higher limit

acceptable (>43µmol/L) with   Gilbert’s syndrome

Trial criteria

No trial data regarding patients outside of these parameters

AST/ALT

≤ 4xULN or trial-specific criteria should be met

Attempt to identify cause of liver   derangement e.g. infection, drug toxicity including antifungals, VoD, GvHD,   disease

Creatinine clearance

> 30 ml/min

Physicians should consider appropriate dose reductions in

cyclophosphamide and fludarabine when creatinine   clearance is below   60ml/min and potentially an increased   interval between LD and CAR-T return to permit clearance of Fludarabine metabolites.

Cardiac function

Repeat cardiac investigations    only if clinically indicated e.g.  clinical signs and symptoms of heart   failure, cardiotoxic

bridging chemotherapy.

Repeat TTE, ECG and cardiac biomarkers (troponin and   NT-proBNP); Cardio-oncology   assessment is required

Assessment of disease burden

Baseline assessment

PET-CT/ other imaging; bone marrow; LP as indicated

Table 7. Checklist before starting LD conditioning. Key. LD: lymphodepletion;  WBC: white blood cell count; ALC: absolute lymphocyte count; ULN: upper limit of normal; VoD: veno-occlusive disease; GvHD: graft versus host disease; TTE: trans-thoracic echography; ECG: electrocardiogram

 

 

PRODUCT RECEIPT

Oversight and responsibility for this process varies nationally. Country-specific guidance is beyond the scope of this document. Centers should have regulatory approval for storage of genetically modified organisms (GMOs).

Transport tracking on the manufacturer’s website enables the date and time of product shipment to be known in advance. At receipt, checks should include: (1) inspection of the dry shipper seal for breaches; (2) review of the temperature log throughout transportation; (3) inspection of product integrity; (4) CAR-T identity label checks, prior to completion of receipt forms. In the  event  of  non-conformance,   cells   are  quarantined,  and  the  hospital   delegate  and manufacturer should be immediately informed. Back-up bags are sometimes available as a replacement for defective products.

 

 

 

Out-of-specification (OOS) CAR-T may still be used (exceptions include microbial/noxious contamination), provided the release certificate lists the OOS details, written agreement is provided from the manufacturer (with acceptance of responsibility by both manufacturer and physician), and the patient has given written consent30-32.

 

 

THAWING AND INFUSION

Journal Pre-proofPrior to thawing and infusion, patients are medically assessed to ensure they are fit to proceed (Table 8); identity and consent is confirmed; the prescription reviewed, and vital signs and IV access (central venous catheter or a newly inserted and pre-tested peripheral cannula) checked at the bedside. Pre-medication with paracetamol and antihistamine (NOT corticosteroids) is recommended. No concurrent medication should be administered during CAR-T infusion.

Product thawing is performed in a pharmacy clean room, cell therapy unit or patient bedside, double wrapped in a watertight plastic bag, using thawing devices according to manufacturer’s instructions and local regulations (automated thawing device, 37±2°C water bath, or dry-thaw method). CAR-T is stable at room temperature for 30-90 minutes after thawing33.

CAR-T should be administered rapidly after thawing during working hours by competent medical or nursing personnel13,14 . Vital signs should be assessed and recorded before, during and following infusion. Using aseptic non-touch technique (ANTT), cells in vials are drawn up into a syringe to be administered as a slow bolus. CAR-T infusion bags are infused through a non-filtered giving set.   Fluid infusion sets with sub-micrometre bacterial filters and blood transfusion sets with leukocyte depletion filters should NOT be used for CAR-T infusion.

Infusion reactions are rare but should be treated symptomatically. Corticosteroids should be avoided unless the patient becomes critically unwell.

Following infusion, the vial/bag and giving set should be disposed of as a GMO biohazard in compliance with institutional policies and country-specific regulations.

 

 

 

Complications

EBMT/EHA

recommendations

Comments

Active infection

Contraindication

CAR T-cell infusion should be delayed until the infection   is controlled

Clinical evidence of fluid   overload or congestive cardiac   failure

Contraindication

Specific individualized risk-benefit; cardio oncology assessment is required

Cardiac arrhythmia not controlled with medical management

Contraindication

Specific individualized risk-benefit; cardio oncology assessment is required

Hypotension requiring vasopressor support

Contraindication: work-up is required to identify cause

CAR T-cell infusion should be delayed until the hypotension is controlled

New-onset or worsening of another non-   hematologic organ dysfunction ≥ Grade 3

Work-up is needed to identify   the cause

Specific individualized risk-benefit assessment required

Significant worsening of clinical condition since   start of LD

Work-up is needed to identify   the cause

Specific individualized risk-benefit assessment required

Neurological evaluation including ICE score (adult) or CAPD score   (children)

To be routinely performed

Serving as a baseline

Table  8. Potential complications to be ruled out before product thawing and CAR-T infusion. Key. LD: lymphodepletion

 

 

 

SHORT-TERM COMPLICATIONS: ADMISSION TO DAY +28

 

Tumour lysis syndrome: TLS can occur following LD/CAR-T, and should be prevented and managed with standard local protocols.

 

 

Infection: Active infection should be controlled prior to initiation of LD.  Following LD, all patients will be neutropenic. A fever should prompt empiric antimicrobial therapy (based on institutional protocols) and investigation with blood and urine cultures; chest x-ray and/or high- resolution CT chest, when indicated; respiratory viral screening including COVID-19 and other tests such as influenza A PCR testing; cytomegalovirus (CMV) and Epstein-Barr virus (EBV) nucleic acid testing (NAT); and lumbar puncture and brain MRI in selected cases. The specific risk profile of the patient (duration of neutropenia, prior allo-HCT, previous infections, and local  antibiotic  resistance  profiles)   should  guide  diagnostic  work-up  and  selection  of antimicrobial agents.

 

 

Cytokine release syndrome (CRS): CRS affects 30-100% of all patients, with ≥grade 3 CRS reported in 10-30%34. Incidence depends on the CAR-T product, disease characteristics and CRS grading system used. Typical onset is between  1-14  days post-CAR-T  infusion and duration is commonly between 1-10 days. Rare delayed cases are reported9.

CRS is characterised by fever 38°C, hemodynamic instability and hypoxemia. Severity is graded according to the ASTCT consensus criteria (Figure 1)35  and the differential diagnosis includes neutropenic sepsis. Empiric, broad-spectrum IV antibiotics should be commenced.

CAR-T activation leads to effector cytokine release (IFN-γ, TNF-α, IL-2) which can trigger pro-inflammatory cytokine release (IL-1, IL-6, IFN- γ, IL-10 and MCP1), with increased CRP and hyperferritinemia24. Risk factors for high-grade CRS include tumor burden, concurrent infection, CAR-T dose and product, and LD conditioning intensity36.

CRS management combines symptomatic measures (antipyretics, fluids) with tocilizumab (IL6 receptor antagonist) +/-corticosteroids10. When two doses of tocilizumab (8mg/kg) fail to control CRS, dexamethasone should be administered. Tocilizumab should be used earlier in older, co-morbid patients. Early/prophylactic tocilizumab has been studied in CRS but there is insufficient data to date to support a formal recommendation of this approach.

Clinicians should be vigilant for occult sepsis emerging post-tocilizumab. Gastrointestinal perforation has also been reported37. Corticosteroids should be subject to rapid taper once CRS is controlled.

If CRS does not respond to tocilizumab/corticosteroids, alternative therapeutic options include siltuximaband anakinra, but limited clinical data is available38. There should additionally be a high  index  of  suspicion   for  underlying/concurrent  infection  or  macrophage  activation syndrome (MAS). A suggested CRS management algorithm is shown in Figure 1.

 

 

 

 

Journal Pre-proofFigure 1: Algorithm outlining the grading and management of CRS

 

 

Macrophage  Activation  Syndrome  (MAS):  Persistent  fever  despite  tocilizumab  with organomegaly,  cytopenias  (+/-  hemophagocytosis  in  the bone marrow), hyperferritinemia (>10,000ng/mL),     liver     dysfunction,     coagulopathy      (hypofibrinogenemia     requiring cryoprecipitate/fibrinogen concentrate) and hypertriglyceridemia, favors a CRS/MAS overlap syndrome rather than CRS39.

Patients should be monitored with twice daily blood tests (WBC, liver function, ferritin, CRP) and treated with anakinra, a recombinant humanised IL-1 receptor antagonist, in combination with corticosteroids40  (Figure 2). In refractory CRS/MAS, chemotherapy can be used, albeit there is  a  lack  of data  and  a  high risk  of ablating CAR-T. Where there is neurological involvement, intrathecal chemotherapy can be considered41,42 .

 

 

 

Journal Pre-proofFigure 2: Management of CRS/MAS (expert opinion based on literature review) 38-42

 

 

Immune effector cell-associated neurotoxicity syndrome (ICANS): ICANS affects 20-60% of CD19CAR-T patients (grade ≥3, 12-30%). Onset is typically three to five days after CAR- T but can occur concurrently with/shortly after CRS, and 10% of patients develop ‘delayed ICANS’ more than three weeks after infusion43,44 . Classical ICANS is also reported, to a lesser extent, with CD22- and BCMA-targeting CAR-T4. On the CARTITUDE study of LCAR- B38M,   a   series   of  movement/neurocognitive   disorders/nerve   palsies/peripheral   motor neuropathies have been observed (12% of cases), not temporally associated with CRS, that are of later onset (median day 27), and take longer to resolve (median 75 days)45. These will require ongoing evaluation in clinical trials. ICANS is rarely reported in solid tumor CAR-T studies.

The pathophysiology of ICANS is likely to be due to a combination of inflammatory cytokines increasing  vascular  permeability;  endothelial  activation  leading  to  blood-brain  barrier breakdown;  increased  CSF  cytokines  and  in  some  cases  leading  to   cerebral  edema40. Pharmacokinetics indicate that greater, earlier CAR-T expansion in vivo correlates with higher ICANS risk46. Risk factors for ≥ grade 3 ICANS include CD28-CAR-Tproducts, higher CAR- T doses, high disease burden, pre-existing neurological conditions, low platelet count, and early, severe CRS47. High fever (≥38.9°C) and hemodynamic instability within 36 hours of CAR-T infusion predicts for severe ICANS with high sensitivity48.

Symptoms include tremor, confusion, agitation, and seizures. Dysphasia, hesitant speech and deterioration  in  handwriting  is  prominent  and  can  progress  to  expressive  and  receptive aphasia48. Routine anti-convulsant prophylaxis is not recommended except in high-risk cases. Fatal cerebral  edema has been described48. Late psychiatric presentations have also been reported49.

ICANS is a clinical diagnosis, but MRI brain and CSF examination can exclude alternative diagnoses10. Electroencephalogram (EEG) can be normal but can also demonstrate slowing and non-convulsive  status  epilepticus.  Diagnostic  work-up  should  include  CT  head,  clotting screen/fibrinogen and EEG, MRI, and lumbar puncture (LP) in severe ICANS, or steroid- refractory cases.

 

 

 

Duration and frequency of ICANS monitoring should be conducted as per product label/trial protocol. The ten-point Immune Effector Cell Encephalopathy (ICE) score in adults (Table 9) and the CAPD assessment in children (Table 10) is usually performed twice daily. ICANS grading integrates the ICE/CAPD scores into an overall assessment of neurological function (Figure 3).

Journal Pre-proofManagement is supportive for grade 1 ICANS. Corticosteroid therapy with a rapid taper is indicated for grade ≥2 ICANS and ICU transfer should be considered (Figure 3). Evidence suggests that steroids do not impact CAR-T efficacy, although longer courses can be associated with  shorter  PFS50.  Seizures  are  treated  with  levetiracetam  and  status  epilepticus  with benzodiazepines. There is no clear therapeutic role for tocilizumab in ICANS and it has been suggested that it may contribute to ICANS through increase circulating IL-646. In the specific setting of grade 1 CRS with concurrent grade 2 ICANS, it is appropriate that steroids (and not tocilizumab) be administered, but this does not apply in higher grade CRS. Alternative agents include siltuximab and anakinra, but clinical data on their utility in ICANS is limited. A management algorithm for ICANS is shown in Figure 3.

 

Test

Points

Orientation: orientation to year,   month, city, hospital

4

Naming: ability to name three objects   (e.g., table, television, pillow)

3

Following commands: ability to follow   simple commands (e.g., “smile” or “open   your mouth”)

1

Writing: ability to write a standard   sentence (e.g., “Happy to have my family around”)

1

Attention: ability to count backwards from 100 by 10

1

Table 9. ICE score for neurological toxicity assessment. Adapted from Lee et al48

 

Test

always

often

sometimes

rarely

never

Eye contact with caregiver

0

1

2

3

4

Purposeful actions

0

1

2

3

4

Aware of their surroundings

0

1

2

3

4

Being restless

4

3

2

1

0

Being inconsolable

4

3

2

1

0

Being underactive

4

3

2

1

0

Slow response to interactions

4

3

2

1

0

Communicating needs and wants

5

4

3

2

40

Table 10. CAPD for encephalopathy assessment in children < 12 years, Adapted from Traubeet al51,52.

 

 

 

Figure 3: Grading and management of ICANs

 

 

Cardiovascular    Toxicity:10-20%     of    CAR-T     patients     experience     cardiovascular complications53. Hypotension requiring vasopressor support was the main cardiac complication observed in pediatric CAR-T studies, but arrhythmias, myocardial impairment, left ventricular systolic dysfunction (LVSD), decompensated cardiac failure, and cardiovascular death are reported54.

Risk factors for CAR-T-cardiotoxicity include grade 2 CRS55, more often in high disease burden  and  patients  with  pre-existing  cardiac   dysfunction  following  prior  exposure  to cardiotoxins including anthracyclines, radiation, and tyrosine kinase inhibitors.

Thorough  pre-CAR-T  cardiovascular  assessment  with  appropriate   surveillance  and  risk reduction  strategies  may  reduce  CAR-T-cardiovascular  complications.  Elevated  baseline serum cardiac biomarkers (troponin, N-terminal-pro-brain-natriuretic-peptide (NT-proBNP), may signal a greater risk of CAR-T-cardiotoxicity. Electrocardiograph (ECG) will exclude underlying   arrhythmias   and   QT   interval    (as   a   surrogate   for   cardiac   repolarisation abnormalities)  and transthoracic  echocardiography  (TTE)  defines baseline  left ventricular ejection  fraction  (LVEF)  and diastolic  function to identify pre-existing LVSD  (using 4D volumetric, 2D Simpson’s Biplane, and global longitudinal strain assessment tools). Cardiac magnetic resonance (CMR) can be considered in the setting of poor image quality, including patients with pericardial/myocardial involvement on PET to assess lymphomatous infiltration.

 

 

 

On admission, baseline ‘dry weight’ and daily weights should be recorded, where weight increase as a surrogate for fluid overload should prompt repeat cardiac assessment with serum biomarkers, ECG, TTE and cardiologist review.

Tocilizumab is also associated with rapid improvement in cardiovascular complications56. One retrospective analysis showed a 1.7-fold increased risk of CAR-T-cardiotoxicity with each 12- hour delay intocilizumab administration from CRS onset55. Current experience suggests that CAR-T-cardiotoxicity is an early, largely reversible phenomenon, with rare LVSD beyond 6 months and no late cardiovascular effects at one year49.

 

 

Journal Pre-proofLaboratory testing:  CRP,  fibrinogen,  liver  function tests  and  ferritin  are  checked  daily. Cytokine  testing  is  not  routinely  performed  in  most  centers.  Atypical  lymphocytes  that resemble   leukemic   blasts    are   not    uncommon   at    peak   CAR-T    expansion.   Repeat microbiological testing and imaging to exclude infection is recommended in febrile patients.

 

 

MEDIUM TERM COMPLICATIONS: DAY +28 TO DAY +100

 

 

Delayed TLS/CRS/ICANS: Although rare, delayed events can occur and should be managed according to standard protocols (Figures  1, 2 and 3). Table 11 outlines recommended testing during this period to monitor for complications.

 

Tests

EBMT/EHA recommendations

Comments

Purpose

Frequency

FBC, Biochemistry panel, AST,

Standard follow-up

At every visit and as

ALT, bilirubin, LDH, Fibrinogen,

clinically indicated

CRP

CMV, EBV, Adenovirus, COVID-19

Viral reactivation/ infection (post-allo-HCT)

As clinically indicated

Quantitative Immunoglobulins or Serum protein   electrophoresis

Immune reconstitution

1-3 monthly

Consider IV (or SC) immunoglobulin replacement

Peripheral blood

Immune recovery

Once monthly for first three

Guide to anti-infective prophylaxis

Immunophenotyping –

months, three monthly

and vaccination schedule

CD3/4/8/16+56/19+

thereafter in first year

CAR T monitoring

CAR T persistence

Peripheral blood flow

cytometry or transgene   by molecular methods as

clinically indicated

This is   not feasible in most centers.   For B-ALL, B-cell aplasia maybe   used as a surrogate for persistence

Table 11. Patient monitoring during medium-term follow-up. Abbreviations. FBC: full blood count; CMV: cytomegalovirus; EBV; Epstein-Barr virus; IV: intravenous; SC: subcutaneous.  * Additional tests and imaging can be performed as clinically indicated.

 

 

Infections  and  antimicrobial  prophylaxis:  Opportunistic  infections  are  common  and prophylaxis  is  warranted  until  immune  reconstitution  (Table   12)43.    Risks  include  prior autologous/allo-HCT, bridging therapy, and steroids/tocilizumab for CRS/ICANS. Prolonged neutropenia, CD4 T-cell lymphopenia and B-cell aplasia/hypogammaglobulinemia (affecting up to 46% of patients at Day +90) also contribute. Neutropenia beyond day +30 and beyond

 

 

 

day +90 affects 30% and 10-20% of patients, respectively. Prolonged CD4 T-cell lymphopenia resolves to >200/µL by 1 and 2 years in 65% and 86% of patients, respectively57.

Most early infections (first 30 days) are bacterial, or respiratory viral infections. Invasive fungal infections are rare. Risk factors include B-ALL with prior allo-HCT; prior fungal infection and prior long-term/high-dose steroid exposure58.

Beyond Day +30, viral infections predominate. HSV and VZV reactivation is uncommon in patients on valaciclovir prophylaxis. CMV, EBV, adenovirus, HHV6, BK-virus and JC-virus reactivation are rare and routine monitoring is not advised, except in high-risk patients (allo- HCT; high-dose/long-term corticosteroids)59. COVID-19 is a formidable challenge in CAR-T patients and consortium guidance on how to maintain CAR-T delivery through the pandemic is available.

Journal Pre-proofEvidence suggests that CAR-T manufacturing is feasible in HBV, HCV and HIV infection and treatment is safe provided the virus is undetectable before apheresis and prior to starting LD. In    hepatitis    B    infection     (especially    if    HBsAg+     and    HBV-DNA+),    long-term entecavir/tenofovir/equivalent prophylaxis is recommended.

 

EBMT/EHA recommendation

Comments

Neutropenia

G-CSF to shorten duration of neutropenia   from day +14 or after resolution of CRS orICANS.

Can consider starting earlier, e.g., day 5*, if patient is at

high risk of infection, e.g., ALL, post-allo-HCT,   high-dose   steroids. For persistent   neutropenia (<0.5 x 109/L) following Day +28, consider G-CSF.

Avoid if patient has CRS   orICANS

Antibacterial prophylaxis

Not routinely recommended**

Can be considered in case of prolonged neutropenia and

should be based on local guidelines e.g., with levofloxacin or ciprofloxacin.

Anti-viral

Valaciclovir 500 mg bid or aciclovir 800mg bid

Start from LD conditioning until one-year post-CAR T- cell infusion AND until CD4count >0.2x109/L

Anti-pneumocystis

Co-trimoxazole 480 mg once daily   or 960 mg three times each week.

To start from LD conditioning until one-year post-CAR-T cell infusion AND until CD4+count >0.2x109/L.

Where there is prolonged myelosuppression, postpone start after   ANC > 0.5 x 109/L.

Can be started later depending on centre guidelines.

In case of co-trimoxazole allergy (or cytopenias

precluding use of co-trimoxazole), pentamidine inhalation   (300 mg once every month), dapsone 100 mg daily or

atovaquone 1500   mg once daily can be considered.

Systemic anti-fungal prophylaxis

Not recommended routinely; consider posaconazole

(300mg/d) or fluconazole (200 mg/d) or micafungin (50 mg i.v./d) inpatients   with severe (ANC < 0.5 x 109/L) or

prolonged (> 14 days)   neutropenia and/or inpatients on

long-term or high dose   (> 72 h) corticosteroids or inpatients post-allo-HCT.

Inpatients with prior allo-HCT, prior invasive

aspergillosis and those receiving corticosteroids, posaconazole prophylaxis should   be considered.

IV Immunoglobulin

Routine in children. Consider in adults with

serious/recurrent infections with encapsulated organisms and hypogammaglobulinemia (<   4g/L).

Clinical evidence does not support routine use in adults following   allo-HCT.

Table 12. Infection prophylaxis post-CAR-T. Abbreviations.  G-CSF: granulocyte colony stimulating factor; CRS:  cytokine  release syndrome: LD:  lymphodepleting  conditioning; NCCN:  The National  Comprehensive Cancer Network, allo-HCT: allogeneic hematopoietic cell transplantation.

* A negative impact of G-CSF applied earlier after CAR-Thas not been clearly demonstrated; a recent report starting G- CSF on day 5 after CAR-T infusion showed no increase in CRS orICANS, indicating that earlier application may be safe and may reduce duration of neutropenia. Further data to support this observation is required.60

**  In  patients  with  neutropenic  fever,   empiric   treatment  with  broad-spectrum  antibiotics  is  strongly recommended.

 

 

 

B-cell aplasia & hypogammaglobulinemia: Following CAR-T, B-cell aplasia is associated with sino-pulmonary infections and should be measured regularly10. 83% of paediatric B-ALL patients on the ELIANA study had ongoing B-cell aplasia at 6 months. In ZUMA-1 responders, 25% had ongoing B-cell aplasia at 12months.

Due to immunological immaturity, immunoglobulin replacement is routine in pediatric CAR- T. In adults, long-lived plasma cells following CD19 CAR-T may confer an immune-protective effect, but a common approach is immunoglobulin replacement for hypogammaglobulinemia (<4g/L) with serious or recurrent/chronic infections. Data on efficacy of immunoglobulin replacement in CAR-T is limited and current recommendations are mainly extrapolated from primary immunodeficiencies (e.g., Bruton’s agammaglobulinemia). One study showed that increased serum IgG led to significantly less sinopulmonary infection post-CAR-T61.

Immunoglobulin replacement aims to maintain serum levels above 4 g/L in adults and within age-adapted normal ranges for children, titrated to the incidence of breakthrough infection.

Intravenous immunoglobulins (IVIGs) (0.4g/kg) and subcutaneous (SCIGs) (0.1–0.15g/kg) are administered 3-6 weekly, and weekly, respectively. After reaching steady state, levels should be measured three monthly.

Cessation of immunoglobulin replacement should be guided by recovery offunctional B-cells. This also provides a surrogate for functional CAR-T persistence and maybe useful in clinical decision making, particularly in B-ALL.

 

 

Vaccinations: General guidance is outlined in Table  13, applicable to adults and children. Incomplete immune reconstitution or ongoing immunosuppression confers a high likelihood of lower vaccine responses (including COVID-19), but consensus view is that vaccination may reduce infection rates and improve clinical outcome. Recommendations and adherence to national schedules require individualized assessment based on infection history and laboratory assessments of cellular/humoral immunity, where available. Specific antibody responses to vaccination should be assessed where possible. A recent analysis in adults following CD19- and BCMA-CAR-T indicated that despite hypogammaglobulinemia, CD19-CAR-T patients developed seroprotection comparable to the general population (with the exception of specific pathogens e.g., pneumococcus), but that in BCMA-CAR-T patients, fewer pathogen-specific antibodies  were   detected62.  This  highlights   the  need   for  vaccine   and  immunoglobulin replacement studies in this at-risk population. Further analysis of T-cell vaccine responses is also warranted in this group.

 

 

Agent

EBMT/EHA recommendations

Comments

Pre-CAR-T

Post-CAR-T

Influenza Vaccine

Preferably vaccinate 2 weeks prior to LD.

In B-cell aplasia low

likelihood of serological response

> 3 months after CAR-T   patients should be

vaccinated irrespective of immunological

reconstitution

Where there is incomplete immune reconstitution* or      ongoing immunosuppression, there is a high likelihood of lower   vaccine responses. Consensus view is that

vaccination may still be beneficial to reduce rates of   infection and improve clinical course.   Consider boost upon B-cell recovery.

SARS-Cov-19

Preferably vaccinate prior to CAR-T therapy.

In B-cell aplasia low

likelihood of serological response

> 3 months after CAR T-cell   infusion.

No reports on vaccine response after CAR-T exist.

However, SARS-Cov-19   vaccine-induced protection    relies heavily on T-cell mediated   immunity, therefore   B-cell aplasia does not seem to be a contraindication;   No T cell   threshold has been defined. Post vaccination response monitoring is desirable. Consider boost upon B-cell recovery.

 

 

 

 

Killed / Inactivated vaccines

> 6 months after CAR-T and   > 2 months after

immunoglobulin

replacement

Contraindications include concurrent

immunosuppressive or cytotoxic therapy.

Live and non-live adjuvant vaccines

1 year after CAR-T and fully immune

reconstituted *

Contraindications include < 2 years post allo-HCT, < 8

months after completion of immunoglobulin replacement.

Table 13. Eligibility Criteria for Vaccination inpatients receiving CD19-targeted CAR T-cell therapy [adapted from Hill and Seo. 63* Absolute  CD4 T cells  > 0.2x109/L, CD19 or CD20 positive B-cells > 0.2x109/L, no concomitant immunosuppressive or cytotoxic therapy

 

 

Graft-versus-Host Disease  (GvHD):  CAR-T post-allo-HCT  is  generally  considered  safe without  increased  risk  of  high-grade  GvHD.  However,  a  recent  publication   describes histologically confirmed GvHD in 4 pediatric patients post-KymriahTM.64  GvHD post-CAR-T should be diagnosed and managed using standard protocols, balancing the benefits of systemic immunosuppression against the adverse impact on CAR-T viability.

 

 

Delayed cytopenias: Haematological toxicity has a cumulative one-year-incidence of 58%, post-CD19-CAR-T, is often prolonged and can follow a biphasic temporal course, with initial neutrophil recovery followed by a ‘second dip’65.

Duration and severity vary between products and indications, but there is a high incidence of persistent grade ≥3 neutropenia (30-38%), thrombocytopenia (21-29%), and anemia (5-17%) after Day +2866-69. Risk factors include baseline cytopenias, pre-treatment bone marrow disease burden, an inflammatory state, prior allo-HCT within one year, and severe CRS/ICANS65,70 . Protracted cytopenias are less pronounced in BCMA- and solid tumor targeting CAR-T.6,68

The pathophysiology remains poorly understood and there may be product-intrinsic and/or disease-specific     factors.      Investigations      should     consider      hematinic      deficiency, myelosuppressive medications (co-trimoxazole), and viral infections (HHV6,Parvovirus B19). Bone  marrow  biopsy  may  be  useful  beyond   day  28  to   exclude   recurrent   disease, hemophagocytosis and rarely, myelodysplasia.

G-CSF can be used for severe neutropenia (<0.5 x 109/L) from day +14 onwards, following resolution of CRS/ICANS. Recent data on earlier prophylactic G-CSF found no effect on immunotoxicity, CAR-T expansion, or prognosis.60

G-CSF-refractory neutropenia (ANC <100/µl) lasting ≥30 days affects 5-10% of patients, with a risk of fungal infection71. Autologous stem cell rescue is an option, where cells are available72 and donor-derived, unconditioned CD34+-selected ‘top-up’ can be considered in post-allo- HCT patients73.  Anti-inflammatory therapies (dexamethasone) and EPO/TPO-agonists may have a role. Allo-HCT is the last resort inpatients with refractory cytopenias.

 

 

LONG-TERM FOLLOW-UP (LTFU): FROM DAY +100

LTFU should be conducted by a multi-disciplinary team (CAR-T physicians; disease-specific specialists; LTFU nursing staff; data managers; clinical trial staff) to capture disease status and late effects14.

Prolonged cytopenias, hypogammaglobulinemia and infections  are common. Neurological complications  and  pulmonary  toxicity  confer  an  increased  mortality  risk.   Secondary malignancy is rare: a single report of relapse following transduction of a leukemicB-cell during the manufacturing process is described74 and a case of myelodysplastic syndrome was reported

 

 

in the ZUMA-1 trial.  A recent publication described CAR-T-derived malignancies following genome-edited CD19-CAR-T due to insertional mutagenesis75. This area requires ongoing surveillance.

CAR-T centres should liaise with referral centres, providing protocols and policies for LTFU, to sustain shared care arrangements.

A recommended LTFU schedule of attendance and testing schedule are outlined in Tables 13 and 14.

 

Period

EBMT/EHA recommendations

Visit frequency

Outcomes to be monitored:

Day +100 to one year

Monthly

 

Journal Pre-proofDisease – remission, minimal residual disease (MRD) status,   relapse, death Subsequent   treatments including allo-HCT and other IEC therapy/ATMP    Immunological   status – immune cell markers, immunoglobulins, CAR T

persistence

New cancers/ secondary myeloid diseases

Autoimmunity and new autoimmune diseases

Endocrine, reproductive and bone health including growth and development Neurological status   (recovery from ICANS)

Psychological status and quality of life

Cardiovascular disease, including risk factors such as   metabolic syndrome Respiratory function

Gastrointestinal and liver health

Vaccination guidance (see Section   3.4)

Patients who proceed to subsequent allo-HCT, cytotoxic   therapy and/or immune effector cell therapy should be followed asper Majhail et al 2012131

One year to two years

Six-monthly

Two to fifteen years

Annually

Table 13. Recommended minimum frequency of attendance at CAR-T centre for patients in remission for Late Effect monitoring.

 

Test

Purpose

EBMT/EHA

recommendations

Comments

Frequency

Full Blood Count,   Biochemistry panel

Standard   follow-up

At   every visit

Viral infection (PB PCR,   NPA)

Viral

reactivation/infection

As clinically indicated

Quantitative   Immunoglobulins +/- serum protein   electrophoresis

Immune

reconstitution

At   every visit

Peripheral blood   Immunophenotyping

– CD3/4/8/16+56/19*

Immune

reconstitution

Every   second visit

No longer required

following   normalization

CAR-T monitoring where commercial

CAR-T   persistence

Every   visit

No longer required

kits are available for   routine monitoring

when   absent for two

of anti-CD19 CAR-T*

consecutive   tests

Endocrine   function and other standard   late effects testing appropriate to age

Standard   follow-up

Yearly or as clinically indicated

Table 14. Recommended tests to be performed at LTFU clinic. Key: PB:peripheral blood; NPA: nasopharyngeal aspiration. *Equivalent test methods for other immune effector cells as they become available.



https://m.sciencenet.cn/blog-3426442-1437741.html

上一篇:[转载]Onasemnogene abeparvovec基因疗法治疗症状性婴儿型脊髓性肌萎缩症1型-part2

0

该博文允许注册用户评论 请点击登录 评论 (0 个评论)

数据加载中...

Archiver|手机版|科学网 ( 京ICP备07017567号-12 )

GMT+8, 2024-6-18 21:58

Powered by ScienceNet.cn

Copyright © 2007- 中国科学报社

返回顶部