Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 + 3699 word(s) 3699 2021-05-24 06:05:27 |
2 format correct -24 word(s) 3675 2021-06-01 03:26:37 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Karschnia, P. CAR T-Cells for CNS Lymphoma. Encyclopedia. Available online: https://encyclopedia.pub/entry/10321 (accessed on 20 April 2024).
Karschnia P. CAR T-Cells for CNS Lymphoma. Encyclopedia. Available at: https://encyclopedia.pub/entry/10321. Accessed April 20, 2024.
Karschnia, Philipp. "CAR T-Cells for CNS Lymphoma" Encyclopedia, https://encyclopedia.pub/entry/10321 (accessed April 20, 2024).
Karschnia, P. (2021, May 31). CAR T-Cells for CNS Lymphoma. In Encyclopedia. https://encyclopedia.pub/entry/10321
Karschnia, Philipp. "CAR T-Cells for CNS Lymphoma." Encyclopedia. Web. 31 May, 2021.
CAR T-Cells for CNS Lymphoma
Edit

Primary or secondary central nervous system (CNS) lymphoma is frequently associated with a poor prognosis. CAR T-cells are being established as a relevant treatment approach in hematological B-cell malignancies. Unfortunately, most clinical studies on chimeric antigen-receptor (CAR) T-cells have excluded patients with CNS involvement but several clinical trials on CAR T-cell therapy in CNS lymphoma patients are currently ongoing. Preclinical and preliminary clinical data suggest an overall acceptable safety profile and considerable anti-tumor effects might be extrapolated for CAR T-cell therapy in CNS lymphoma. 

refractory relapsed central nervous system adoptive survival immunotherapy

1. Introduction

Primary central nervous system lymphoma (PCNSL) represent a rare group of extranodal B-cell non-Hodgkin lymphomas arising from the brain parenchyma, spinal cord, eyes, or meninges without systemic, extra-axial involvement [1]. Such tumors account for 2% of all primary central brain tumors [2][3]. Antimetabolites including methotrexate and cytarabine represent the backbone of anti-PCNSL therapy, and may be followed by consolidation radiotherapy or high dose chemotherapy and autologous stem cell transplantation [4]. The addition of chemotherapy to the former standalone radiotherapy has translated into substantially improved survival [5]; however, PCNSL is still associated with limited outcome compared to extra-axial disease and a median survival of less than three years [6]. Importantly, radiotherapy is frequently accompanied by disabling neurotoxicity including decline in cognitive function, and such effects need to be carefully weighed against potential benefits in terms of survival [7]. Secondary CNS lymphomas refer to secondary involvement of the neuroaxis by systemic disease, and often indicate aggressive disease with unfavorable survival compared to systemic disease only [8]. Median survival after diagnosis of secondary CNS lymphoma is only about four months [8]. The identification of new therapeutic approaches for primary and secondary CNS lymphomas is therefore urgently warranted.
Adoptive immunotherapy with chimeric antigen receptor (CAR) T-cells has emerged as an efficient therapy for relapsed or refractory hematological malignancies [9]. Following viral transduction, CARs direct the killing properties of an autologous T-cell population against a tumor cell antigen. To increase persistence, activity, and expansion, CARs are equipped with a costimulatory domain [10]. Numerous clinical studies demonstrated substantial response rates for CAR T-cells directed against the pan-B cell antigen CD19 in patients with diffuse large B-cell lymphoma [11], B-cell acute lymphoblastic leukemia [12], and mantle-cell lymphoma [13]. Five CAR T-cell products are currently available for commercial use in the United States and the European Union and constitute a major breakthrough in the treatment of hematological cancers. Given that almost all CNS lymphoma manifestations express CD19 [14][15], there is a strong biologic rationale to treat such patients with CD19-directed CAR T-cells. However, there is no definitive conclusion on whether this indeed represents a promising therapeutic avenue. We herein provided a review on the available literature for CAR T-cells in the treatment of PCNSL and also secondary CNS lymphoma. We summarized recent preclinical and clinical data on CAR T-cell therapy for primary and secondary CNS lymphoma, discussed challenges when treating primary brain tumors with CAR T-cells, and hypothesized on future directions of the field.

2. Preclinical and Clinical Data

2.1. Preclinical Data

Anti-tumor effects of CD19-directed CAR T-cells against PCNSL have not only been demonstrated in vitro, but also in murine in vivo models [16][17]. Mulazzani et al. designed an orthotopic PCNSL model by combining a chronic cranial window with two-photon intravital microscopy, allowing the repetitive visualization of brain tumor growth [16]. A single dose of intracerebrally injected CD19-directed CAR T-cells was not only able to mediate regression, but also to completely eliminate established PCNSL in two out of three animals. These substantial anti-tumor effects lasted up to half a year until experiments were terminated, and CAR T-cells resided in the brain parenchyma as well as in draining and non-draining lymph nodes throughout the observation period. Importantly, intravenous CAR T-cell injection was associated with a low number of tumor-infiltrating CAR T-cells and therefore not able to sufficiently control PCNSL growth. Although the authors speculated that this might be due to poor trafficking of CAR T-cells across the blood–brain barrier or a rather low number of intravenously injected cells, the final mechanisms behind this observation were not elucidated. Importantly, the study was limited by the use of immunoincompetent mice lacking functional T-cells (but retain B-cells) as human lymphoma cells were utilized.
PCNSL regression after local but not intravenous administration of CAR T-cells was recently corroborated in another immunoincompetent mouse model (lacking function T- and B-cells) of PCNSL [18]. Wang et al. induced orthotopic PCNSL growth by intracranial injection of human lymphoma cell lines. CD19-directed CAR T-cells were either delivered via a single intraventricular or intravenous infusion. Bioluminescence was measured to quantify tumor growth in vivo over the course of weeks, and only intraventricularly injected CAR T-cells were able to control PCNSL growth. Single-cell RNA analysis of CAR T-cells sampled from the bone marrow of post-treatment mice, in vitro culture of CAR T-cells in either cerebrospinal fluid (CSF) or medium, and further mechanistic analyses showed that exposure to the CSF results in a distinct anti-tumor and memory effectivity of CAR T-cells.
These findings made from preclinical studies appear therefore promising in controlling PCNSL; however, they have not yet been validated in immunocompetent animal models. Given that only a limited number of preclinical studies on CAR T-cells and PCNSL is available, a high level of suspicion is therefore required when interpreting these results; however, some anti-tumor effects against PCNSL and CNS lymphoma in general might be assumed.

2.2. Clinical Data

Patients with active involvement of the brain were excluded from almost all clinical trials on CAR T-cells, mainly due to dreaded more severe neurotoxic side effects. These trials have resulted in US Food and Drug Administration (FDA) approval of CD19-directed CAR T-cells for patients with systemic but not CNS disease [11][12]. So far, only three studies analyzing the clinical efficiency of CAR T-cells in patients with primary or secondary CNS lymphoma are available to date, all of them using CAR T-cells targeting CD19 [19][20][21] (Table 1).
Table 1. Published studies on CAR T-cells for treatment of primary and secondary CNS lymphoma.
  Study Design Study Population Route of Delivery Antigens Toxicities Outcome NCT/
ChiCTR
Abramson et al. [19] Case report on a patient enrolled in a phase 1 clinical trial
  • Secondary CNS lymphoma (n = 1):
  • Diffuse large B-cell lymphoma
Intravenously Lisocabtagene maraleucel (formerly JCAR017):
CD19CAR T-cells
None CR after 1 months NCT02631044
Frigault et al. [20] Retrospective cohort study
  • Secondary CNS lymphoma (n = 8):
  • Diffuse large B-cell lymphoma (n = 5)
  • High-grade B-cell lymphoma (n = 2)
  • Primary mediastinal B-cell lymphoma (n = 1)
Intravenously Tisagenlecleucel:
CD19CAR T-cells
  • Grade 1 CRS (n = 7)
  • No NT
  • No tocilizumab or steroid treatment needed
  • PD (n = 4) with † on day 3 and 25 (n = 2)
  • PR (n = 2) with ongoing control on day 90 (n = 1) and 180 (n = 1)
  • CR (n = 2) with ongoing control on day 90 (n = 1) 180 (n = 1)
NCT04134117
Siddiqi et al. [21] Preliminary data from an ongoing phase 1 clinical trial
  • Primary CNS lymphoma (n = 3)
  • Secondary CNS lymphoma (n = 4)
  • Intravenously (n = 7)
  • Intraventricular, under evaluation
CD19CAR T-cells modified to express a truncated eGFR
  • Grade 1–2 NT and CRS, treated with steroids (n = 2) or tocilizumab (n = 3)
  • CR (n = 1)
  • PR (n = 3)
NCT02153580
Li et al. [22] Phase 1 clinical trial
  • Primary CNS lymphoma (n = 1)
  • Secondary CNS lymphoma (n = 4)
Intravenously Combination of:
  • CD19CAR T-cells
  • CD22CAR T-cells
  • Grade 1 (n = 4) and 2 (n = 1) CRS
  • Grade 1 (n = 1) and 4 (n = 1) NT, treated with steroids, plasmapheresis, tocilizumab
60-days assessment:
  • CR (n = 1)
  • PR (n = 4)
ChiCTR-OPN-16008526
In 2017, a first case report on CAR T-cell efficacy in secondary CNS lymphoma was published [19]. Abramson et al. enrolled a 68-year-old female with refractory diffuse large B-cell lymphoma in the TRANSCEND-NHL-001 trial on the CAR T-cell product lisocabtagene maraleucel (formerly known as JCAR017). After T-cell apheresis and prior to lymphodepletion and CAR T-cell infusion, re-staging studies were provided and a new right temporal mass consistent with disease involvement of the CNS was noted on imaging. The patient proceeded with lymphodepletion and intravenous CAR T-cell infusion (NCT02631044) as initially planned, and complete remission of the cerebral lymphoma site was seen one month after infusion. Of note, this remission was durable and ongoing for 12 months at the time the report was published. Neither cytokine release syndrome nor neurotoxicity was noted.
Another CD19-directed CAR T-cell product, tisagenlecleucel (formerly known as CTL019), has been approved in 2017 for large B-cell lymphoma patients with systemic but also secondary (not primary) CNS involvement. Based on the granted FDA approval, Frigault et al. treated and reported on a retrospective cohort of eight patients with secondary CNS involvement of the brain, spine, and leptomeninges [20]. All patients received lymphodepletion and a single intravenous CAR T-cell infusion of tisagenlecleucel (0.6 × 108 to 6.0 × 108 CAR T-cells). Only mild neurotoxic or systemic side effects were encountered, and none of these patients experienced CAR T-cell-mediated toxicities necessitating therapy with the anti-interleukin 6-receptor antagonist tocilizumab or steroids. Response assessment on day 28 after CAR T-cell infusion showed complete response in two patients, partial response in two more patients, and disease progression in four patients (including two fatalities due to progressive disease). Further follow-up on day 90 revealed ongoing disease control in three of the four patients who initially responded to CAR T-cells, and long-term follow up on day 180 was available in one of those patients showing complete response.
These results suggesting considerable anti-tumor effects in the treatment of CNS disease were recently corroborated by preliminary data from an ongoing prospective trial of CD19-directed CAR T-cells for B-cell non-Hodgkin lymphoma (NCT02153580) [21]. The studied CAR T-cell product is modified to express a truncated human epidermal growth factor receptor, which may serve as an antibody target to rapidly eliminate CAR T-cells in vivo in case of severe toxicities. Three patients with primary and four patients with secondary CNS lymphoma were treated by intravenous CAR T-cell infusion (2 × 108 to 6 × 108 CAR T-cells) following lymphodepletion, whereas when no life-threatening toxicities occurred, tocilizumab was provided for moderate cytokine release syndrome in two patients and steroids for neurotoxicity in three patients. Four patients had disease responses to CAR T-cells with one patient showing complete response and three patients showing partial response. On a cautionary note, follow-up time was only in the range of several weeks and it is unclear whether this response was durable.
Data from longer follow-up intervals after treatment of CNS disease were reported from Li et al. (ChiCTR-OPN-16008526) [22]. One patient with primary and four patients with secondary CNS lymphoma each received one intravenous infusion of CD19-directed (2.2 × 106 to 7.1 × 106/kg body weight) and one infusion of CD22-directed CAR T-cells (3.1 × 106 to 7.0 × 106/kg). CD22 is another pan B-cell marker which offers an additional target in the case of CD19 antigen loss [23]. In this cohort, one case of mild neurotoxic symptoms and one case of high-grade neurotoxicity was encountered which necessitated the use of steroids and plasmapheresis. All 5 patients responded within 60 days after CAR T-cell administration including two complete responses. However, four patients relapsed within three to eight months, and median progression-free survival was three months. Despite tumor relapse, tumor tissue analysis and CSF studies in one patient showed persistent target antigen expression and detectable CAR T-cells. The authors speculated that the immunosuppressive tumor microenvironment providing resistance against CAR T-cells might have contributed to tumor recurrence. However, the authors lacked sufficient evidence for this theory.
Based on the encouraging results of above-mentioned studies, different clinical phase I and phase II trials are currently testing safety and efficiency of CD19 CAR T-cells in primary and secondary CNS lymphoma patients (Table 2).
Table 2. Current clinical trials to primary and secondary CNS lymphoma.
Sponsor Study Chair Study Design Population Conditions Interventions Route of Application NCT
University College London Claire Roddie Phase I clinical trial Adults (>16 years)
  • Refractory/relapsed primary CNS lymphoma
Anti-CD19 CAR T-cells after lymphodepletion and pembrolizumab
  • Intravenously
  • Intraventricularly via Ommaya reservoir
NCT04443829
Massachusetts General Hospital Matthew J. Frigault Phase I clinical trial Adults (>18 years)
  • Refractory/relapsed primary CNS lymphoma
Tisagenlecleucel (anti-CD19 CAR T-cells after lymphodepletion) Intravenously NCT04134117
Dana-Farber Cancer Institute Caron A. Jacobson Phase I clinical trial Adults (>18 years)
  • Refractory/relapsed central nervous system (CNS) lymphoma
  • Systemic lymphoma with concurrent CNS lymphoma
Axicabtagene ciloleucel (anti-CD19 CAR T-cells after lymphodepletion) Intravenously NCT04608487
Memorial Sloan Kettering Cancer Center Jae Park Phase I dose-escalation trial Adults (>18 years)
  • Refractory/relapsed central nervous system (CNS) lymphoma
  • Systemic lymphoma with concurrent CNS lymphoma
Anti-CD19 19(T2)28z1XX CAR T-cells Intravenously NCT04464200
Celgene Claudia Schusterbauer Phase II clinical trial Adults (>18 years)
  • Refractory/relapsed central nervous system (CNS) lymphoma
  • Systemic lymphoma with concurrent CNS lymphoma
Lisocabtagene maraleucel (anti-CD19 CAR T-cells after lymphodepletion) Intravenously NCT03484702
Zhejiang University He Huang Early phase I clinical trial
  • Children (>3 years)
  • Adults (18–75 years)
  • Acute lymphoblastic leukemia with CNS involvement
  • Non-Hodgkin’s lymphoma with CNS involvement
Anti-CD19 CAR T-cells after lymphodepletion Intraventricularly NCT04532203

3. Challenges for CAR T-Cells in CNS Lymphoma

The above-mentioned studies suggest an acceptable safety profile of CAR T-cells for CNS lymphoma. Furthermore, considerable anti-tumor effects have been reported. Whether these anti-tumor effects are as long-lasting and profound as it has been described for extracranial disease might be in doubt. A number of CNS-specific aspects may hamper clinical success of such therapies.

3.1. Immune-Escaping Tumor Properties

Primary brain tumors including PCNSL represent complex compositions of neoplastic and non-neoplastic cells which individually contribute to tumor formation [24]. Tumor-associated macrophages and microglia (TAM/M) constitute the majority of non-neoplastic cells in PCNSL, and these cells create a particular immunosuppressive pre-metastatic niche which facilitates tumor cell extravasation, survival, and expansion [25]. A spectrum of TAM/M activation phenotypes have been defined between the pro-inflammatory, anti-tumor M1 and the anti-inflammatory, pro-tumor M2 phenotype [26]. Accordingly, higher numbers TAM/M polarized towards M2 phenotype are associated with less favorable outcome in PCNSL [27]. In addition, immunosuppressive cytokines are strongly expressed in PCNSL, whereas cytokines promoting cell-based immune response are downregulated [28]. To mitigate the immunosuppressive milieu, different approaches including CAR T-cells expressing inducible proinflammatory cytokines [29] or combination therapies with checkpoint inhibitors [30] are currently being investigated in preclinical trials.

3.2. Role of the Blood–Brain Barrier and Route of CAR T-Cell Application

In addition to metabolic barriers for tumor infiltration by CAR T-cells, physical barriers including the blood–brain barrier may limit treatment success. Under physiologic conditions, the brain is virtually free of leucocytes, and their influx is tightly regulated. CAR T-cells have been shown to migrate across the blood–brain barrier and can be found in brain and CSF [16][31]. In turn, locally injected CAR T-cells have not only been found to travel to distant sites within the CSF but can also be detected in the systemic circulation [16][32]. After intravenous injection, the number of CAR T-cells within the CSF seems generally lower than in the systemic circulation [31]. In preclinical models, intravenous administration of CAR T-cells for CNS lymphoma but also other brain tumors such as glioblastoma has provided considerable anti-tumor effects [31][33]. However, direct comparison of different routes of application indicate that local delivery may be associated with improved treatment response in brain tumors [16][34]. Importantly, immunodeficient PCNSL mouse models may lack proper function of circulating B-cells and CD19-directed CAR T-cells may therefore not encounter their target immediately after intravenous injection (in contrast to local delivery). This may impair CAR T-cell expansion, and thus underestimate their anti-tumor effectivity. However, insufficient anti-tumor effects after intravenous injection of CAR T-cells have not only been observed in the murine model by Wang et al. [18] who used NOD scid gamma mice (lacking B- and T-cell function), but also in the murine model by Mulazzani et al. [16] who made use of Foxn1nu/nu mice (lacking T- but not B-cell function). However, the murine CD19 on B-cells from Foxn1nu/nu mice differs substantially from human CD19 which the CAR T-cells were directed against in the study by Mulazzani et al. It might therefore be indeed speculated that these models may underestimate CAR T-cell effectivity.
Clinical studies for glioblastoma show that local CAR T-cell delivery (delivery into a resection cavity or administration into the CSF) is feasible and effective [16][18][34]. Several clinical studies using local delivery of CAR T-cells to treat primary brain tumors other than PCNSL are currently recruiting (NCT03500991, NCT03638167, NCT04185038). Local injection of CAR T-cells for CNS lymphoma has so far not yet been conducted; however, it might represent an approach warranting evaluation especially for patients with primary CNS lymphoma given the exclusive CNS involvement.

3.3. Antigen Loss

Another factor contributing to recurrence after CAR T-cell therapy may be the loss or downregulation of the tumoral target antigens. Of patients with B-cell leukemia, 7–25% experience CD19-negative relapse [12][35]. The frequency of antigen loss in lymphoma is less clear given that biopsies are rarely obtained during relapse. However, several of such cases have been described after CAR T-cell therapy for systemic lymphoma [36], and antigen loss might therefore be also relevant for PCNSL [37]. In the future, (re-)biopsies of cerebral manifestations should be encouraged for antigenic profiling of the new lesion in order to substantiate the presence of druggable targets. Approaches to prevent or circumvent antigen loss as potential escape mechanism will also need to be evaluated in CNS lymphoma.

3.4. Adverse Effects of CAR T-Cells

CAR T-cell therapy directed against CD19, but also other antigens might be accompanied by unique toxicities including cytokine release syndrome (CRS), immune effector cell-associated neurotoxicity syndrome (ICANS), on-target–off-tumor toxicities, and prolonged cytopenia [38][39].
CRS represents the most commonly encountered adverse effect and is characterized by a systemic increase of pro-inflammatory cytokines translating into sepsis-like symptoms [40]. A high number of up to 93% of patients with extra-axial lymphoma treated with CAR T-cells may experience some degree of CRS, and one out of ten patients may experience severe symptoms necessitating treatment at an intensive care unit [38]. So far, in the treatment of CNS lymphoma, only mild cases of CRS were seen with a frequency similar to what has been observed for systemic disease [11][12][21].
ICANS is the second most commonly observed toxicity following CAR T-cell therapy. Clinical presentation varies and includes moderate symptoms such as headaches, fatigue, or aphasia [41], but also more severe and potentially life-threatening symptoms such as seizures/status epilepticus, cerebral edema, and death [42]. Pathophysiology is likely multifactorial and involves IL-1- and IL-6-mediated systemic inflammation, blood–brain barrier disruption, endothelial activation, and cross-reactivity of CAR T-cells against brain tissue [43][44]. On-target–off-tumor toxicity refers to effects caused by CAR T-cells against non-pathogenic tissue due to shared expression of target antigens on neoplastic and healthy tissue. Parker et al. recently demonstrated by single-cell RNA sequencing and autopsy studies that brain mural cells, which are critical for blood–brain barrier integrity, express CD19 [44]. Thus, an on-target mechanism may contribute to the development of ICANS. The occurrence of severe ICANS has been associated with decreased survival after CAR T-cell therapy [45]. Treatment consists in the application of steroids. Tocilizumab, which can be used to treat CRS, does not seem to improve ICANS. As therapy escalation plasmapheresis, the application of immunoglobulins, and the IL1-antagonist anakinra may show beneficial effects in individual cases [46][47][48]. Although there has been major concern that treatment of CNS disease may be paralleled by strong neurotoxic symptoms, only one case of severe ICANS in a CNS lymphoma patient receiving CD19 and CD22 directed CAR T-cells has so far been described [22]. In clinical trials investigating CAR T-cell therapy for other primary brain malignancies like glioblastoma, only few cases with severe CRS and no clinical manifestation of ICANS were reported [49]. No data exist for locally injected CAR T-cells in CNS lymphoma therapy which would also circumvent direct exposition of brain mural cells to CAR T-cells, and future clinical trials will need to closely monitor for ICANS. In addition, long-term effects of CAR T-cell treatment on cognitive performance have been described [50], and the heavy pre-treatment burden including whole-brain radiotherapy of CNS lymphoma patients may aggravate such effects [7].
Hematological toxicities are among the most common, yet underreported adverse effects of CAR T-cell therapy [39]. As all commercially available CAR T-cell products are exclusively approved for patients with refractory or relapsed malignancies, all patients underwent extensive treatment prior to CAR T-cell infusion which frequently results in profound and long-lasting cytopenia. Moreover, lymphodepletion is usually used prior to CAR T-cell infusion as it may enhance anti-tumor efficacy by decreasing regulatory T-cells and myeloid-derived suppressor cells, increasing levels of proinflammatory cytokines, and enhancing innate immunity [51]. Such an approach is also often associated with profound cytopenia; however, the extent and frequency of observed cytopenia cannot fully be explained by lymphodepletion or prior chemotherapies alone and likely also includes CAR T-cell-mediated mechanisms [52]. A high level of suspicion is required as CAR T-cell patients are therefore highly susceptible for viral, bacterial, or fungal infections [53]. Problematically, therapeutic agents which alleviate cytopenia such as GM-CSF could potentially worsen other toxicities such as ICANS [54]. One could argue that local delivery of CAR T-cells (e.g., intratumoral injection of CAR T-cells or intraventricular application via indwelling ventricle catheter) holds the potential to decrease systemic adverse effects like cytopenia by decreasing circulating CAR T-cells in the blood stream; however, data on that subject remain scarce and future clinical trials will have to address different routes of administration for PCNSL immunotherapy.

3.5. Hematological Limitations: Lymphopenia and Autoimmune Diseases

Whereas patients with glioblastoma and primary brain tumors other than PCNSL often experience lymphopenia before, during, and after CAR T-cell therapy as stated above, patients with CNS lymphoma are at a particularly high risk due to aggressive myeloablative first-line therapies which also include the use of stem cell transplantation [55][56][57]. In selected patients with low lymphocyte counts, apheresis of an adequate quantity of autologous T-cells for CAR T-cell manufacturing might be challenging. Fortunately, with a continuous improvement in lymphapheresis protocols, sufficient yields of lymphocytes might be expected in most patients [58]. Of note, CAR T-cells from aged donors are of impaired quality including shorter persistence and less memory-like phenotypes and one may speculate that there might also be an association with more extensive pre-treatment burden [59].
Moreover, autoimmune disorders requiring immunosuppressive medication are overrepresented among patients with CNS lymphoma [60]. The pathomechanistic implications of this observations are not fully understood but likely involve Epstein–Barr virus-induced mutations and such patients might be at particular risk for less favorable outcome [61][62]. Importantly, individuals with autoimmune disease were excluded from the landmark clinical trials that resulted in the approval of commercial CAR T-cell products, and there is only little evidence whether CAR T-cell therapy may aggravate symptoms or not [63]. It remains to be shown whether CAR T-cell therapy is safe and beneficial also in patients with CNS lymphoma and a history of autoimmune disease.

References

  1. Karschnia, P.; Batchelor, T.T.; Jordan, J.T.; Shaw, B.; Winter, S.F.; Barbiero, F.J.; Kaulen, L.D.; Thon, N.; Tonn, J.-C.; Huttner, A.J.; et al. Primary Dural Lymphomas: Clinical Presentation, Management, and Outcome. Cancer 2020, 126, 2811–2820.
  2. Ostrom, Q.T.; Gittleman, H.; Truitt, G.; Boscia, A.; Kruchko, C.; Barnholtz-Sloan, J.S. CBTRUS Statistical Report: Primary Brain and Other Central Nervous System Tumors Diagnosed in the United States in 2011–2015. Neuro Oncol. 2018, 20, iv1–iv86.
  3. Han, C.H.; Batchelor, T.T. Diagnosis and Management of Primary Central Nervous System Lymphoma. Cancer 2017, 123, 4314–4324.
  4. Ferreri, A.J.M. How I Treat Primary CNS Lymphoma. Blood 2011, 118, 510–522.
  5. Glass, J.; Gruber, M.L.; Cher, L.; Hochberg, F.H. Preirradiation Methotrexate Chemotherapy of Primary Central Nervous System Lymphoma: Long-Term Outcome. J. Neurosurg. 1994, 81, 188–195.
  6. Houillier, C.; Soussain, C.; Ghesquières, H.; Soubeyran, P.; Chinot, O.; Taillandier, L.; Lamy, T.; Choquet, S.; Ahle, G.; Damaj, G.; et al. Management and Outcome of Primary CNS Lymphoma in the Modern Era: An LOC Network Study. Neurology 2020, 94, e1027–e1039.
  7. Karschnia, P.; Parsons, M.W.; Dietrich, J. Pharmacologic Management of Cognitive Impairment Induced by Cancer Therapy. Lancet Oncol. 2019, 20, e92–e102.
  8. El-Galaly, T.C.; Cheah, C.Y.; Bendtsen, M.D.; Nowakowski, G.S.; Kansara, R.; Savage, K.J.; Connors, J.M.; Sehn, L.H.; Goldschmidt, N.; Shaulov, A.; et al. Treatment Strategies, Outcomes and Prognostic Factors in 291 Patients with Secondary CNS Involvement by Diffuse Large B-Cell Lymphoma. Eur. J. Cancer 2018, 93, 57–68.
  9. June, C.H.; Sadelain, M. Chimeric Antigen Receptor Therapy. N. Engl. J. Med. 2018, 379, 64–73.
  10. Savoldo, B.; Ramos, C.A.; Liu, E.; Mims, M.P.; Keating, M.J.; Carrum, G.; Kamble, R.T.; Bollard, C.M.; Gee, A.P.; Mei, Z.; et al. CD28 Costimulation Improves Expansion and Persistence of Chimeric Antigen Receptor–Modified T Cells in Lymphoma Patients. J. Clin. Investig. 2011, 121, 1822–1826.
  11. Schuster, S.J.; Bishop, M.R.; Tam, C.S.; Waller, E.K.; Borchmann, P.; McGuirk, J.P.; Jäger, U.; Jaglowski, S.; Andreadis, C.; Westin, J.R.; et al. Tisagenlecleucel in Adult Relapsed or Refractory Diffuse Large B-Cell Lymphoma. N. Engl. J. Med. 2019, 380, 45–56.
  12. Maude, S.L.; Laetsch, T.W.; Buechner, J.; Rives, S.; Boyer, M.; Bittencourt, H.; Bader, P.; Verneris, M.R.; Stefanski, H.E.; Myers, G.D.; et al. Tisagenlecleucel in Children and Young Adults with B-Cell Lymphoblastic Leukemia. N. Engl. J. Med. 2018, 378, 439–448.
  13. Wang, M.; Munoz, J.; Goy, A.; Locke, F.L.; Jacobson, C.A.; Hill, B.T.; Timmerman, J.M.; Holmes, H.; Jaglowski, S.; Flinn, I.W.; et al. KTE-X19 CAR T-Cell Therapy in Relapsed or Refractory Mantle-Cell Lymphoma. N. Engl. J. Med. 2020, 382, 1331–1342.
  14. Deckert, M.; Montesinos-Rongen, M.; Brunn, A.; Siebert, R. Systems Biology of Primary CNS Lymphoma: From Genetic Aberrations to Modeling in Mice. Acta Neuropathol. 2014, 127, 175–188.
  15. Giannini, C.; Dogan, A.; Salomão, D.R. CNS Lymphoma: A Practical Diagnostic Approach. J. Neuropathol. Exp. Neurol. 2014, 73, 478–494.
  16. Mulazzani, M.; Fräßle, S.P.; von Mücke-Heim, I.; Langer, S.; Zhou, X.; Ishikawa-Ankerhold, H.; Leube, J.; Zhang, W.; Dötsch, S.; Svec, M.; et al. Long-Term in Vivo Microscopy of CAR T Cell Dynamics during Eradication of CNS Lymphoma in Mice. Proc. Natl. Acad. Sci. USA 2019, 116, 24275–24284.
  17. Hu, S.-I.; Ko, M.-C.; Dai, Y.-H.; Lin, H.-A.; Chen, L.-C.; Huang, K.-Y.; Pang, T.-L.; Kuo, C.-Y.; Lin, H.-C. Pre-Clinical Assessment of Chimeric Antigen Receptor t Cell Therapy Targeting CD19+ B Cell Malignancy. Ann. Transl. Med. 2020, 8, 584.
  18. Wang, X.; Huynh, C.; Urak, R.; Weng, L.; Walter, M.; Lim, L.; Vyas, V.; Chang, W.-C.; Aguilar, B.; Brito, A.; et al. The Cerebroventricular Environment Modifies CAR T Cells for Potent Activity against Both Central Nervous System and Systemic Lymphoma. Cancer Immunol. Res. 2021, 9, 75–88.
  19. Abramson, J.S.; McGree, B.; Noyes, S.; Plummer, S.; Wong, C.; Chen, Y.-B.; Palmer, E.; Albertson, T.; Ferry, J.A.; Arrillaga-Romany, I.C. Anti-CD19 CAR T Cells in CNS Diffuse Large-B-Cell Lymphoma. N. Engl. J. Med. 2017, 377, 783–784.
  20. Frigault, M.J.; Dietrich, J.; Martinez-Lage, M.; Leick, M.; Choi, B.D.; DeFilipp, Z.; Chen, Y.-B.; Abramson, J.; Crombie, J.; Armand, P.; et al. Tisagenlecleucel CAR T-Cell Therapy in Secondary CNS Lymphoma. Blood 2019, 134, 860–866.
  21. Siddiqi, T.; Wang, X.; Palmer, J.; Popplewell, L.L.; Nikolaenko, L.; Herrera, A.F.; Budde, L.E.; Lim, L.; Vyas, V.; Brown, C.E.; et al. CD19-Targeting CAR-T Cell Therapy in CNS Lymphoma. Blood 2019, 134, 4075.
  22. Li, T.; Zhao, L.; Zhang, Y.; Xiao, Y.; Wang, D.; Huang, L.; Ma, L.; Chen, L.; Liu, S.; Long, X.; et al. CAR T-Cell Therapy Is Effective but Not Long-Lasting in B-Cell Lymphoma of the Brain. Front. Oncol. 2020, 10.
  23. Shah, N.N.; Highfill, S.L.; Shalabi, H.; Yates, B.; Jin, J.; Wolters, P.L.; Ombrello, A.; Steinberg, S.M.; Martin, S.; Delbrook, C.; et al. CD4/CD8 T-Cell Selection Affects Chimeric Antigen Receptor (CAR) T-Cell Potency and Toxicity: Updated Results From a Phase I Anti-CD22 CAR T-Cell Trial. J. Clin. Oncol. 2020, 38, 1938–1950.
  24. Sampson, J.H.; Gunn, M.D.; Fecci, P.E.; Ashley, D.M. Brain Immunology and Immunotherapy in Brain Tumours. Nat. Rev. Cancer 2020, 20, 12–25.
  25. Hambardzumyan, D.; Gutmann, D.H.; Kettenmann, H. The Role of Microglia and Macrophages in Glioma Maintenance and Progression. Nat. Neurosci. 2016, 19, 20–27.
  26. Murray, P.J.; Allen, J.E.; Biswas, S.K.; Fisher, E.A.; Gilroy, D.W.; Goerdt, S.; Gordon, S.; Hamilton, J.A.; Ivashkiv, L.B.; Lawrence, T.; et al. Macrophage Activation and Polarization: Nomenclature and Experimental Guidelines. Immunity 2014, 41, 14–20.
  27. Nam, S.J.; Kim, S.; Kwon, D.; Kim, H.; Kim, S.; Lee, E.; Kim, T.M.; Heo, D.S.; Park, S.H.; Lim, M.S.; et al. Prognostic Implications of Tumor-Infiltrating Macrophages, M2 Macrophages, Regulatory T-Cells, and Indoleamine 2,3-Dioxygenase-Positive Cells in Primary Diffuse Large B-Cell Lymphoma of the Central Nervous System. Oncoimmunology 2018, 7, e1442164.
  28. Korfel, A.; Schlegel, U. Diagnosis and Treatment of Primary CNS Lymphoma. Nat. Rev. Neurol. 2013, 9, 317–327.
  29. Chmielewski, M.; Kopecky, C.; Hombach, A.A.; Abken, H. IL-12 Release by Engineered T Cells Expressing Chimeric Antigen Receptors Can Effectively Muster an Antigen-Independent Macrophage Response on Tumor Cells That Have Shut down Tumor Antigen Expression. Cancer Res. 2011, 71, 5697–5706.
  30. Shen, S.H.; Woroniecka, K.; Barbour, A.B.; Fecci, P.E.; Sanchez-Perez, L.; Sampson, J.H. CAR T Cells and Checkpoint Inhibition for the Treatment of Glioblastoma. Exp. Opin. Biol. Ther. 2020, 20, 579–591.
  31. Santomasso, B.D.; Park, J.H.; Salloum, D.; Riviere, I.; Flynn, J.; Mead, E.; Halton, E.; Wang, X.; Senechal, B.; Purdon, T.; et al. Clinical and Biological Correlates of Neurotoxicity Associated with CAR T-Cell Therapy in Patients with B-Cell Acute Lymphoblastic Leukemia. Cancer Discov. 2018, 8, 958–971.
  32. Keu, K.V.; Witney, T.H.; Yaghoubi, S.; Rosenberg, J.; Kurien, A.; Magnusson, R.; Williams, J.; Habte, F.; Wagner, J.R.; Forman, S.; et al. Reporter Gene Imaging of Targeted T Cell Immunotherapy in Recurrent Glioma. Sci. Transl. Med. 2017, 9.
  33. Sampson, J.H.; Choi, B.D.; Sanchez-Perez, L.; Suryadevara, C.M.; Snyder, D.J.; Flores, C.T.; Schmittling, R.J.; Nair, S.K.; Reap, E.A.; Norberg, P.K.; et al. EGFRvIII MCAR-Modified T-Cell Therapy Cures Mice with Established Intracerebral Glioma and Generates Host Immunity against Tumor-Antigen Loss. Clin. Cancer Res. 2014, 20, 972–984.
  34. Brown, C.E.; Alizadeh, D.; Starr, R.; Weng, L.; Wagner, J.R.; Naranjo, A.; Ostberg, J.R.; Blanchard, M.S.; Kilpatrick, J.; Simpson, J.; et al. Regression of Glioblastoma after Chimeric Antigen Receptor T-Cell Therapy. N. Engl. J. Med. 2016, 375, 2561–2569.
  35. Turtle, C.J.; Hanafi, L.-A.; Berger, C.; Gooley, T.A.; Cherian, S.; Hudecek, M.; Sommermeyer, D.; Melville, K.; Pender, B.; Budiarto, T.M.; et al. CD19 CAR-T Cells of Defined CD4+:CD8+ Composition in Adult B Cell ALL Patients. J. Clin. Investig. 2016, 126, 2123–2138.
  36. Zhang, Z.; Chen, X.; Tian, Y.; Li, F.; Zhao, X.; Liu, J.; Yao, C.; Zhang, Y. Point Mutation in CD19 Facilitates Immune Escape of B Cell Lymphoma from CAR-T Cell Therapy. J. Immunother. Cancer 2020, 8.
  37. Nayyar, N.; White, M.D.; Gill, C.M.; Lastrapes, M.; Bertalan, M.; Kaplan, A.; D’Andrea, M.R.; Bihun, I.; Kaneb, A.; Dietrich, J.; et al. MYD88 L265P Mutation and CDKN2A Loss Are Early Mutational Events in Primary Central Nervous System Diffuse Large B-Cell Lymphomas. Blood Adv. 2019, 3, 375–383.
  38. Neelapu, S.S.; Tummala, S.; Kebriaei, P.; Wierda, W.; Gutierrez, C.; Locke, F.L.; Komanduri, K.V.; Lin, Y.; Jain, N.; Daver, N.; et al. Chimeric Antigen Receptor T-Cell Therapy—Assessment and Management of Toxicities. Nat. Rev. Clin. Oncol. 2018, 15, 47–62.
  39. Wudhikarn, K.; Pennisi, M.; Garcia-Recio, M.; Flynn, J.R.; Afuye, A.; Silverberg, M.L.; Maloy, M.A.; Devlin, S.M.; Batlevi, C.L.; Shah, G.L.; et al. DLBCL Patients Treated with CD19 CAR T Cells Experience a High Burden of Organ Toxicities but Low Nonrelapse Mortality. Blood Adv. 2020, 4, 3024–3033.
  40. Fajgenbaum, D.C.; June, C.H. Cytokine Storm. N. Engl. J. Med. 2020, 383, 2255–2273.
  41. Sokolov, E.; Karschnia, P.; Benjamin, R.; Hadden, R.D.M.; Elwes, R.C.D.; Drummond, L.; Amin, D.; Paiva, V.; Pennisi, A.; Herlopian, A.; et al. Language Dysfunction-Associated EEG Findings in Patients with CAR-T Related Neurotoxicity. BMJ Neurol. Open 2020, 2, e000054.
  42. Karschnia, P.; Strübing, F.; Teske, N.; Blumenberg, V.; Bücklein, V.L.; Schmidt, C.; Schöberl, F.; Dimitriadis, K.; Forbrig, R.; Stemmler, H.-J.; et al. Clinicopathologic Findings in Fatal Neurotoxicity After Adoptive Immunotherapy With CD19-Directed CAR T-Cells. Hemasphere 2021, 5, e533.
  43. Norelli, M.; Camisa, B.; Barbiera, G.; Falcone, L.; Purevdorj, A.; Genua, M.; Sanvito, F.; Ponzoni, M.; Doglioni, C.; Cristofori, P.; et al. Monocyte-Derived IL-1 and IL-6 Are Differentially Required for Cytokine-Release Syndrome and Neurotoxicity Due to CAR T Cells. Nat. Med. 2018, 24, 739–748.
  44. Parker, K.R.; Migliorini, D.; Perkey, E.; Yost, K.E.; Bhaduri, A.; Bagga, P.; Haris, M.; Wilson, N.E.; Liu, F.; Gabunia, K.; et al. Single-Cell Analyses Identify Brain Mural Cells Expressing CD19 as Potential Off-Tumor Targets for CAR-T Immunotherapies. Cell 2020, 183, 126–142.e17.
  45. Karschnia, P.; Jordan, J.T.; Forst, D.A.; Arrillaga-Romany, I.C.; Batchelor, T.T.; Baehring, J.M.; Clement, N.F.; Gonzalez Castro, L.N.; Herlopian, A.; Maus, M.V.; et al. Clinical Presentation, Management, and Biomarkers of Neurotoxicity after Adoptive Immunotherapy with CAR T Cells. Blood 2019, 133, 2212–2221.
  46. Xiao, X.; He, X.; Li, Q.; Zhang, H.; Meng, J.; Jiang, Y.; Deng, Q.; Zhao, M. Plasma Exchange Can Be an Alternative Therapeutic Modality for Severe Cytokine Release Syndrome after Chimeric Antigen Receptor-T Cell Infusion: A Case Report. Clin. Cancer Res. 2019, 25, 29–34.
  47. Strati, P.; Ahmed, S.; Kebriaei, P.; Nastoupil, L.J.; Claussen, C.M.; Watson, G.; Horowitz, S.B.; Brown, A.R.T.; Do, B.; Rodriguez, M.A.; et al. Clinical Efficacy of Anakinra to Mitigate CAR T-Cell Therapy-Associated Toxicity in Large B-Cell Lymphoma. Blood Adv. 2020, 4, 3123–3127.
  48. Hill, J.A.; Giralt, S.; Torgerson, T.R.; Lazarus, H.M. CAR-T—and a Side Order of IgG, to Go?—Immunoglobulin Replacement in Patients Receiving CAR-T Cell Therapy. Blood Rev. 2019, 38, 100596.
  49. Goff, S.L.; Morgan, R.A.; Yang, J.C.; Sherry, R.M.; Robbins, P.F.; Restifo, N.P.; Feldman, S.A.; Lu, Y.-C.; Lu, L.; Zheng, Z.; et al. Pilot Trial of Adoptive Transfer of Chimeric Antigen Receptor-Transduced T Cells Targeting EGFRvIII in Patients With Glioblastoma. J. Immunother. 2019, 42, 126–135.
  50. Ruark, J.; Mullane, E.; Cleary, N.; Cordeiro, A.; Bezerra, E.D.; Wu, V.; Voutsinas, J.; Shaw, B.E.; Flynn, K.E.; Lee, S.J.; et al. Patient-Reported Neuropsychiatric Outcomes of Long-Term Survivors after Chimeric Antigen Receptor T Cell Therapy. Biol. Blood Marrow Transplant. 2020, 26, 34–43.
  51. Restifo, N.P.; Dudley, M.E.; Rosenberg, S.A. Adoptive Immunotherapy for Cancer: Harnessing the T Cell Response. Nat. Rev. Immunol. 2012, 12, 269–281.
  52. Jain, T.; Knezevic, A.; Pennisi, M.; Chen, Y.; Ruiz, J.D.; Purdon, T.J.; Devlin, S.M.; Smith, M.; Shah, G.L.; Halton, E.; et al. Hematopoietic Recovery in Patients Receiving Chimeric Antigen Receptor T-Cell Therapy for Hematologic Malignancies. Blood Adv. 2020, 4, 3776–3787.
  53. Rejeski, K.; Kunz, W.G.; Rudelius, M.; Bücklein, V.; Blumenberg, V.; Schmidt, C.; Karschnia, P.; Schöberl, F.; Dimitriadis, K.; von Baumgarten, L.; et al. Severe Candida Glabrata Pancolitis and Fatal Aspergillus Fumigatus Pulmonary Infection in the Setting of Bone Marrow Aplasia after CD19-Directed CAR T-Cell Therapy—A Case Report. BMC Infect. Dis. 2021, 21, 121.
  54. Sterner, R.M.; Sakemura, R.; Cox, M.J.; Yang, N.; Khadka, R.H.; Forsman, C.L.; Hansen, M.J.; Jin, F.; Ayasoufi, K.; Hefazi, M.; et al. GM-CSF Inhibition Reduces Cytokine Release Syndrome and Neuroinflammation but Enhances CAR-T Cell Function in Xenografts. Blood 2019, 133, 697–709.
  55. Ferreri, A.J.M.; Illerhaus, G. The Role of Autologous Stem Cell Transplantation in Primary Central Nervous System Lymphoma. Blood 2016, 127, 1642–1649.
  56. Illerhaus, G.; Kasenda, B.; Ihorst, G.; Egerer, G.; Lamprecht, M.; Keller, U.; Wolf, H.-H.; Hirt, C.; Stilgenbauer, S.; Binder, M.; et al. High-Dose Chemotherapy with Autologous Haemopoietic Stem Cell Transplantation for Newly Diagnosed Primary CNS Lymphoma: A Prospective, Single-Arm, Phase 2 Trial. Lancet Haematol. 2016, 3, e388–e397.
  57. Kasenda, B.; Ihorst, G.; Schroers, R.; Korfel, A.; Schmidt-Wolf, I.; Egerer, G.; von Baumgarten, L.; Röth, A.; Bloehdorn, J.; Möhle, R.; et al. High-Dose Chemotherapy with Autologous Haematopoietic Stem Cell Support for Relapsed or Refractory Primary CNS Lymphoma: A Prospective Multicentre Trial by the German Cooperative PCNSL Study Group. Leukemia 2017, 31, 2623–2629.
  58. Korell, F.; Laier, S.; Sauer, S.; Veelken, K.; Hennemann, H.; Schubert, M.-L.; Sauer, T.; Pavel, P.; Mueller-Tidow, C.; Dreger, P.; et al. Current Challenges in Providing Good Leukapheresis Products for Manufacturing of CAR-T Cells for Patients with Relapsed/Refractory NHL or ALL. Cells 2020, 9, 1225.
  59. Kotani, H.; Li, G.; Yao, J.; Mesa, T.E.; Chen, J.; Boucher, J.C.; Yoder, S.J.; Zhou, J.; Davila, M.L. Aged CAR T Cells Exhibit Enhanced Cytotoxicity and Effector Function but Shorter Persistence and Less Memory-like Phenotypes. Blood 2018, 132, 2047.
  60. Kaulen, L.D.; Karschnia, P.; Dietrich, J.; Baehring, J.M. Autoimmune Disease-Related Primary CNS Lymphoma: Systematic Review and Meta-Analysis. J. Neurooncol. 2020, 149, 153–159.
  61. Kaulen, L.D.; Erson-Omay, E.Z.; Henegariu, O.; Karschnia, P.; Huttner, A.; Günel, M.; Baehring, J.M. Exome Sequencing Identifies SLIT2 Variants in Primary CNS Lymphoma. Br. J. Haematol. 2021.
  62. Kaulen, L.D.; Galluzzo, D.; Hui, P.; Barbiero, F.; Karschnia, P.; Huttner, A.; Fulbright, R.; Baehring, J.M. Prognostic Markers for Immunodeficiency-Associated Primary Central Nervous System Lymphoma. J. Neurooncol. 2019, 144, 107–115.
  63. Jhaveri, K.S.; Schlam, I.; Holtzman, N.G.; Peravali, M.; Richardson, P.K.; Dahiya, S.; Malkovska, V.; Rapoport, A.P. Safety and Efficacy of CAR T Cells in a Patient with Lymphoma and a Coexisting Autoimmune Neuropathy. Blood Adv. 2020, 4, 6019–6022.
More
Information
Subjects: Oncology
Contributor MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register :
View Times: 471
Revisions: 2 times (View History)
Update Date: 01 Jun 2021
1000/1000