CAR - t细胞治疗玻璃体视网膜淋巴瘤的初步证据:一项LOC网络研究

IF 14.6 2区 医学 Q1 HEMATOLOGY
HemaSphere Pub Date : 2025-08-25 DOI:10.1002/hem3.70208
Cécile Pivert, Adélaïde Toutée, Christophe Parizot, Denis Malaise, Alexandre Matet, Valérie Touitou, Magali Le Garff-Tavernier, Damien Roos-Weil, Sarah Touhami, Nabih Azar, Ines Boussen, Véronique Morel, Madalina Uzunov, Clotilde Bravetti, Carole Metz, Eve Todesco, Delphine Sterlin, Carole Soussain, Kahina Ouis Tarhi, Anne Besançon, Khê Hoang-Xuan, Sylvain Choquet, Caroline Houillier, Marine Baron
{"title":"CAR - t细胞治疗玻璃体视网膜淋巴瘤的初步证据:一项LOC网络研究","authors":"Cécile Pivert,&nbsp;Adélaïde Toutée,&nbsp;Christophe Parizot,&nbsp;Denis Malaise,&nbsp;Alexandre Matet,&nbsp;Valérie Touitou,&nbsp;Magali Le Garff-Tavernier,&nbsp;Damien Roos-Weil,&nbsp;Sarah Touhami,&nbsp;Nabih Azar,&nbsp;Ines Boussen,&nbsp;Véronique Morel,&nbsp;Madalina Uzunov,&nbsp;Clotilde Bravetti,&nbsp;Carole Metz,&nbsp;Eve Todesco,&nbsp;Delphine Sterlin,&nbsp;Carole Soussain,&nbsp;Kahina Ouis Tarhi,&nbsp;Anne Besançon,&nbsp;Khê Hoang-Xuan,&nbsp;Sylvain Choquet,&nbsp;Caroline Houillier,&nbsp;Marine Baron","doi":"10.1002/hem3.70208","DOIUrl":null,"url":null,"abstract":"<p>Vitreoretinal lymphoma (VRL) is a rare subtype of large B-cell lymphoma (LBCL) that may present as an isolated condition or with central nervous system (CNS) or systemic involvement. Like the brain, the eye is an immune-privileged site, posing unique therapeutic challenges according to the blood–eye barrier. Despite its typically indolent course, VRL is difficult to eradicate, and the long-term prognosis is poor because of the high risk of CNS involvement.<span><sup>1</sup></span> The median overall survival (OS) is 36–75 months for primary VRL and approximately 57 months for isolated vitreoretinal relapses of primary CNS lymphomas.<span><sup>2</sup></span> Therefore, new therapeutic strategies are urgently needed.</p><p>Anti-CD19 chimeric antigen receptor (CAR) T-cells represent a major advancement in managing systemic LBCL.<span><sup>3, 4</sup></span> Studies show that CAR T-cells can cross the blood–brain barrier and demonstrate high efficacy in CNS lymphomas,<span><sup>5-8</sup></span> but their potential in VRL treatment is not fully understood, and their efficiency has never been described from an ophthalmologic point of view with the newest validated exams.</p><p>Since 2020, CAR T-cell therapy has been considered within the French oculocerebral lymphoma network (LOC network) for relapsed or refractory VRL. This study retrospectively analyzed patients with primary or secondary VRL treated with CAR T-cells.</p><p>We retrospectively identified all VRL patients treated with commercial anti-CD19 CAR T-cells at Pitié-Salpêtrière Hospital (Paris) via the LOC network database until November 2023. Eligible patients had active vitreoretinal involvement, either isolated or with CNS involvement. VRL could be primary or secondary to systemic or CNS lymphoma. Data were retrospectively collected from medical records between April and October 2024. The LOC database was approved by the Institutional Ethical Committee of the coordinating center and the French “Commission Nationale de l'Informatique et des Libertés” (CNIL). All patients provided written informed consent. Responses were evaluated using International Primary CNS Lymphoma Collaborative Group (IPCG) criteria,<span><sup>9</sup></span> and all ophthalmologic examinations of the patient performed immediately before and after the CAR T-cells were systematically reviewed by a single ophthalmologist from the Pitié-Salpétrière Hospital, based on the photographs taken at each consultation (Supplementary Methods). Progression-free survival (PFS), ophthalmic-free survival (OFS), and OS were defined as the time from CAR T-cell infusion: “to CNS, ophthalmic or systemic disease progression or relapse/to ophthalmic progression or relapse/and to death respectively.” Survival rates were calculated using the Kaplan–Meier method. Statistical analyses were conducted using GraphPad Prism v10.0. Cytokine release syndrome and immune effector cell-associated neurotoxicity syndrome (ICANS) were graded per 2019 American Society for Transplantation and Cellular Therapy (ASTCT) guidelines.<span><sup>10</sup></span></p><p>Interleukin (IL)-10 and IL-6 assays were performed on the aqueous humor (AH) using a sensitive Cytometric Bead Array kit (BD Biosciences) on a FACSCanto II cytometer. The AH IL-10 level was classified as detectable (≥2.5 pg/mL) or undetectable (&lt;2.5 pg/mL) based on the laboratory's standard threshold. The presence of CAR T-cells in the blood and AH was measured by flow cytometry. Each sample was incubated for 15 min with 1 µL of CD19 CAR Detection Reagent (Miltenyi Biotec), followed by staining with the following antibodies: anti-Biotin-PE (Miltenyi Biotec), CD45-KrO (clone J33), CD3-Alexa Fluor A750 (clone UCHT1), CD4-ECD (clone SFCI12T4D11), and CD8-Alexa Fluor 700 (clone B9.11). The final sample was analyzed on a DxFLEX flow cytometer (Beckman Coulter), and data were processed using CytExpert (Beckman Coulter) software version 2.0.2.18.</p><p>Between August 2020 and December 2023, seven patients with VRL were treated with CAR T-cells. Their main baseline characteristics and individual outcomes are summarized in Tables 1 and S1.</p><p>At the time of the CAR T-cell therapy decision, four patients had isolated VRL, whereas three also had brain parenchymal involvement. Their median age was 70 years (range, 50–76), and their median Eastern Cooperative Oncology Group performance status was 1 (range, 1–3). The median number of prior therapies before leukapheresis was 4 (range, 2–6), including autologous stem cell transplantation (ASCT) (thiotepa-based induction regimen) in four patients (two patients were ineligible for ASCT at the time of CAR T-cells). All but one patient received bridging therapy using drug regimens directed against ocular/brain disease such as methotrexate, temozolomide, ibrutinib, or cytotoxic chemotherapies (ifosfamide, etoposide, and carboplatin) (Table 1). There were no steroid-containing regimens.</p><p>At the time of CAR T-cell infusion, with regard to ophthalmic involvement, two patients had a complete response (CR), two had a partial response (PR), two had stable disease, and one had progressive disease. Additionally, three patients had CNS progressive disease.</p><p>Three patients received tisagenlecleucel and four received axicabtagene ciloleucel (axi-cel). At 1 month, an ophthalmologic response was observed in all five evaluable patients, including PR in two (40%) and CR in three (60%). Data were unavailable for two patients who developed severe neurotoxicity and could not undergo ophthalmologic assessment at this time (Table S1). During follow-up, the best response achieved was CR or CR unconfirmed for all seven patients. The median time from CAR T-cell infusion to first response was 1 month (range, 1–4). After a median follow-up of 23 months (range, 10–47), the 2-year OFS, PFS, and OS rates were 83%, 69% and 83%, respectively (Figure 1A,B). IL-10 dosage in the AH in six tested patients during the first 6 months post-CAR T-cell therapy was undetectable for all but one patient who relapsed a few months later (Figure S1). On the other hand, IL-6 levels exceeding 2.5 pg/mL were detected in the AH of all six patients tested during this period (range, 19–194 pg/mL), with no observed correlation to the occurrence of grade ≥ 3 ICANS. Two of seven patients experienced relapses: one patient had an isolated intraocular relapse 10 months after CAR T-cell infusion, and the other one experienced a relapse in the brain and cerebrospinal fluid 5 months after CAR T-cell infusion.</p><p>To better characterize the trafficking and persistence of CAR T-cells in the eye, flow cytometry was performed at different time points in three patients (one patient was assessed four times, and two were assessed twice). One patient demonstrated AH positivity for CAR T-cells at 11 days, 1 month, 4 months, and 10 months (last ophthalmologic follow-up) (Figure 1C). The other two patients tested negative for CAR T-cells at all time points. All three patients remained in CR at their last ophthalmologic follow-up (17, 16, and 10 months after CAR T-cell infusion, respectively). Peripheral blood CAR T-cell expansion during the first 30 days post-infusion is illustrated in Figure S2.</p><p>All patients developed cytokine release syndrome (Grade 1 in three patients, Grade 2 in four patients). ICANS of any grade occurred in six patients (Grade 3 in one, Grade 4 in two). The Grade 4 ICANS cases involved a 65-year-old patient with a history of cerebral involvement and whole-brain radiotherapy who received axi-cel for isolated ocular relapse, and a 71-year-old patient who received axi-cel with progressive bulky cerebral disease at the time of infusion. Four out of seven patients received steroids, and no patients developed ocular pseudoprogression.</p><p>This is the first series on CAR T-cell therapy in patients with VRL, and we acknowledge the small and heterogeneous series (VRL both primary and secondary, with or without CNS involvement), which may affect the generalizability of our findings. In particular, it is conceivable that the presence of occult systemic disease, especially in cases of previous systemic involvement, may have facilitated CAR T-cell expansion or that distinct molecular profiles may have influenced the duration of the therapeutic response.</p><p>To date, only a few case reports on CAR T-cell therapy for hematological malignancies with ocular involvement have been published.<span><sup>11-13</sup></span> Taher et al. reported a case of early ocular failure of CAR T-cells in a patient with diffuse large B-cell lymphoma (DLBCL) treated for intraocular and CNS localizations, where the CNS disease remained in CR.<span><sup>14</sup></span> Additionally, two cases of CAR T-cell therapy for B-cell acute lymphoblastic leukemia with ophthalmic localization have been described: one demonstrated persistent CR after 1 year, whereas the other reported a flare reaction with sudden ocular deterioration following CAR T-cell therapy.<span><sup>12, 13</sup></span> The flare reaction consisted of bilateral retinal detachment occurring on Day 7 after CAR T-cell infusion, in the absence of blasts in the AH, but with the presence of white blood cells, 35% of which expressing CD3.</p><p>Although our patients were highly pretreated and refractory, the efficacy of CAR T-cells appeared very promising, with outcomes comparable to those achieved with ASCT within our network (2-year PFS and OS rates of 68% and 87%, respectively).<span><sup>15</sup></span> These results also compared favorably to those obtained in the first-line treatment of primary vitreoretinal lymphoma.<span><sup>1</sup></span> The impact of bridging therapy and the remission status at the time of CAR T-cell infusion may represent confounding factors, but the persistence of response after CAR T-cell infusion in the three highly refractory patients with PR before CAR T-cells (23, 16, and 17 months, respectively) strongly supports the major contribution of CAR T-cells in maintaining disease control. Notably, one patient in CR at 6 months exhibited high IL-10 in the AH (1246 pg/mL) without any clinical signs of relapse but subsequently relapsed at 10 months. The IPCG criteria for ophthalmic response do not account for IL-10 quantification; however, the relapse might have already been present in an infra-clinical state as early as 6 months.</p><p>AH CAR T-cell trafficking was demonstrated for the first time in this series in one patient evaluated at multiple time points, confirming their ability to migrate and persist in the AH despite low blood levels of the target antigen. We can hypothesize that CAR T-cells were detected in the AH of only one of three tested patients due to several factors, including the exceptional localizations in the AH of VRL, technical challenges associated with analyzing small-volume samples, sensitivity of flow cytometry, and potentially suboptimal sampling timing relative to the kinetics of CAR T-cell expansion. Further investigations are needed to better characterize CAR T-cell trafficking in these scenarios.</p><p>Finally, we noted unusual Grade 3–4 ICANS in th1ree of seven patients, while the rate and severity of neurotoxicity reported in CNS lymphomas are comparable to those in systemic lymphomas.<span><sup>5, 7, 16</sup></span> This unexpectedly high rate of severe ICANS in our cohort may be attributed to multiple factors, including the small sample size, a high number of prior treatment lines, a history of ASCT and/or encephalic radiotherapy (both known to be associated with cognitive impairment), the presence of CNS disease (reflecting tumor burden), and the type of CAR T-cell construct used. Notably, among the three patients who developed Grade 3-4 ICANS in our cohort, all had progressive CNS disease; two had previously received ASCT with or without encephalic radiotherapy and were treated with axi-cel. Conversely, we did note any ocular immune-related adverse events during follow-up.</p><p>Larger studies are needed to evaluate the true risk of severe neurotoxicity following CAR T-cell therapy in VRLs. Nevertheless, the decision to pursue CAR T-cell therapy should carefully consider this risk.</p><p>In conclusion, this study is the first to report a significant response rate after CAR T-cell therapy in highly pretreated and refractory VRLs, with a promising 2-year PFS rate of 69%. Longer follow-up is essential to confirm these findings and define the role of CAR T-cell therapy within the broader therapeutic strategy for VRL.</p><p><b>Cécile Pivert</b>: Investigation; formal analysis; writing—original draft; software. <b>Adélaïde Toutée</b>: Investigation; data curation. <b>Christophe Parizot</b>: Formal analysis; investigation; data curation. <b>Denis Malaise</b>: Investigation. <b>Alexandre Matet</b>: Investigation. <b>Valérie Touitou</b>: Investigation. <b>Magali Le Garff-Tavernier</b>: Formal analysis; data curation. <b>Damien Roos-Weil</b>: Investigation. <b>Sarah Touhami</b>: Investigation. <b>Nabih Azar</b>: Investigation. <b>Ines Boussen</b>: Investigation. <b>Véronique Morel</b>: Investigation. <b>Madalina Uzunov</b>: Investigation. <b>Clotilde Bravetti</b>: Formal analysis; data curation. <b>Carole Metz</b>: Investigation. <b>Eve Todesco</b>: Formal analysis; data curation. <b>Delphine Sterlin</b>: Formal analysis. <b>Carole Soussain</b>: Investigation. <b>Kahina Ouis Tarhi</b>: Investigation. <b>Anne Besançon</b>: Investigation. <b>Khê Hoang-Xuan</b>: Investigation. <b>Sylvain Choquet</b>: Conceptualization; supervision; writing—review and editing. <b>Caroline Houillier</b>: Conceptualization; supervision; writing—review and editing; formal analysis; validation; methodology; software. <b>Marine Baron</b>: Conceptualization; validation; supervision; writing—review and editing; methodology; software.</p><p>The authors declare no conflicts of interest.</p><p>No funding.</p>","PeriodicalId":12982,"journal":{"name":"HemaSphere","volume":"9 8","pages":""},"PeriodicalIF":14.6000,"publicationDate":"2025-08-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/hem3.70208","citationCount":"0","resultStr":"{\"title\":\"Preliminary evidence of CAR T-cell therapy activity in vitreoretinal lymphomas: An LOC network study\",\"authors\":\"Cécile Pivert,&nbsp;Adélaïde Toutée,&nbsp;Christophe Parizot,&nbsp;Denis Malaise,&nbsp;Alexandre Matet,&nbsp;Valérie Touitou,&nbsp;Magali Le Garff-Tavernier,&nbsp;Damien Roos-Weil,&nbsp;Sarah Touhami,&nbsp;Nabih Azar,&nbsp;Ines Boussen,&nbsp;Véronique Morel,&nbsp;Madalina Uzunov,&nbsp;Clotilde Bravetti,&nbsp;Carole Metz,&nbsp;Eve Todesco,&nbsp;Delphine Sterlin,&nbsp;Carole Soussain,&nbsp;Kahina Ouis Tarhi,&nbsp;Anne Besançon,&nbsp;Khê Hoang-Xuan,&nbsp;Sylvain Choquet,&nbsp;Caroline Houillier,&nbsp;Marine Baron\",\"doi\":\"10.1002/hem3.70208\",\"DOIUrl\":null,\"url\":null,\"abstract\":\"<p>Vitreoretinal lymphoma (VRL) is a rare subtype of large B-cell lymphoma (LBCL) that may present as an isolated condition or with central nervous system (CNS) or systemic involvement. Like the brain, the eye is an immune-privileged site, posing unique therapeutic challenges according to the blood–eye barrier. Despite its typically indolent course, VRL is difficult to eradicate, and the long-term prognosis is poor because of the high risk of CNS involvement.<span><sup>1</sup></span> The median overall survival (OS) is 36–75 months for primary VRL and approximately 57 months for isolated vitreoretinal relapses of primary CNS lymphomas.<span><sup>2</sup></span> Therefore, new therapeutic strategies are urgently needed.</p><p>Anti-CD19 chimeric antigen receptor (CAR) T-cells represent a major advancement in managing systemic LBCL.<span><sup>3, 4</sup></span> Studies show that CAR T-cells can cross the blood–brain barrier and demonstrate high efficacy in CNS lymphomas,<span><sup>5-8</sup></span> but their potential in VRL treatment is not fully understood, and their efficiency has never been described from an ophthalmologic point of view with the newest validated exams.</p><p>Since 2020, CAR T-cell therapy has been considered within the French oculocerebral lymphoma network (LOC network) for relapsed or refractory VRL. This study retrospectively analyzed patients with primary or secondary VRL treated with CAR T-cells.</p><p>We retrospectively identified all VRL patients treated with commercial anti-CD19 CAR T-cells at Pitié-Salpêtrière Hospital (Paris) via the LOC network database until November 2023. Eligible patients had active vitreoretinal involvement, either isolated or with CNS involvement. VRL could be primary or secondary to systemic or CNS lymphoma. Data were retrospectively collected from medical records between April and October 2024. The LOC database was approved by the Institutional Ethical Committee of the coordinating center and the French “Commission Nationale de l'Informatique et des Libertés” (CNIL). All patients provided written informed consent. Responses were evaluated using International Primary CNS Lymphoma Collaborative Group (IPCG) criteria,<span><sup>9</sup></span> and all ophthalmologic examinations of the patient performed immediately before and after the CAR T-cells were systematically reviewed by a single ophthalmologist from the Pitié-Salpétrière Hospital, based on the photographs taken at each consultation (Supplementary Methods). Progression-free survival (PFS), ophthalmic-free survival (OFS), and OS were defined as the time from CAR T-cell infusion: “to CNS, ophthalmic or systemic disease progression or relapse/to ophthalmic progression or relapse/and to death respectively.” Survival rates were calculated using the Kaplan–Meier method. Statistical analyses were conducted using GraphPad Prism v10.0. Cytokine release syndrome and immune effector cell-associated neurotoxicity syndrome (ICANS) were graded per 2019 American Society for Transplantation and Cellular Therapy (ASTCT) guidelines.<span><sup>10</sup></span></p><p>Interleukin (IL)-10 and IL-6 assays were performed on the aqueous humor (AH) using a sensitive Cytometric Bead Array kit (BD Biosciences) on a FACSCanto II cytometer. The AH IL-10 level was classified as detectable (≥2.5 pg/mL) or undetectable (&lt;2.5 pg/mL) based on the laboratory's standard threshold. The presence of CAR T-cells in the blood and AH was measured by flow cytometry. Each sample was incubated for 15 min with 1 µL of CD19 CAR Detection Reagent (Miltenyi Biotec), followed by staining with the following antibodies: anti-Biotin-PE (Miltenyi Biotec), CD45-KrO (clone J33), CD3-Alexa Fluor A750 (clone UCHT1), CD4-ECD (clone SFCI12T4D11), and CD8-Alexa Fluor 700 (clone B9.11). The final sample was analyzed on a DxFLEX flow cytometer (Beckman Coulter), and data were processed using CytExpert (Beckman Coulter) software version 2.0.2.18.</p><p>Between August 2020 and December 2023, seven patients with VRL were treated with CAR T-cells. Their main baseline characteristics and individual outcomes are summarized in Tables 1 and S1.</p><p>At the time of the CAR T-cell therapy decision, four patients had isolated VRL, whereas three also had brain parenchymal involvement. Their median age was 70 years (range, 50–76), and their median Eastern Cooperative Oncology Group performance status was 1 (range, 1–3). The median number of prior therapies before leukapheresis was 4 (range, 2–6), including autologous stem cell transplantation (ASCT) (thiotepa-based induction regimen) in four patients (two patients were ineligible for ASCT at the time of CAR T-cells). All but one patient received bridging therapy using drug regimens directed against ocular/brain disease such as methotrexate, temozolomide, ibrutinib, or cytotoxic chemotherapies (ifosfamide, etoposide, and carboplatin) (Table 1). There were no steroid-containing regimens.</p><p>At the time of CAR T-cell infusion, with regard to ophthalmic involvement, two patients had a complete response (CR), two had a partial response (PR), two had stable disease, and one had progressive disease. Additionally, three patients had CNS progressive disease.</p><p>Three patients received tisagenlecleucel and four received axicabtagene ciloleucel (axi-cel). At 1 month, an ophthalmologic response was observed in all five evaluable patients, including PR in two (40%) and CR in three (60%). Data were unavailable for two patients who developed severe neurotoxicity and could not undergo ophthalmologic assessment at this time (Table S1). During follow-up, the best response achieved was CR or CR unconfirmed for all seven patients. The median time from CAR T-cell infusion to first response was 1 month (range, 1–4). After a median follow-up of 23 months (range, 10–47), the 2-year OFS, PFS, and OS rates were 83%, 69% and 83%, respectively (Figure 1A,B). IL-10 dosage in the AH in six tested patients during the first 6 months post-CAR T-cell therapy was undetectable for all but one patient who relapsed a few months later (Figure S1). On the other hand, IL-6 levels exceeding 2.5 pg/mL were detected in the AH of all six patients tested during this period (range, 19–194 pg/mL), with no observed correlation to the occurrence of grade ≥ 3 ICANS. Two of seven patients experienced relapses: one patient had an isolated intraocular relapse 10 months after CAR T-cell infusion, and the other one experienced a relapse in the brain and cerebrospinal fluid 5 months after CAR T-cell infusion.</p><p>To better characterize the trafficking and persistence of CAR T-cells in the eye, flow cytometry was performed at different time points in three patients (one patient was assessed four times, and two were assessed twice). One patient demonstrated AH positivity for CAR T-cells at 11 days, 1 month, 4 months, and 10 months (last ophthalmologic follow-up) (Figure 1C). The other two patients tested negative for CAR T-cells at all time points. All three patients remained in CR at their last ophthalmologic follow-up (17, 16, and 10 months after CAR T-cell infusion, respectively). Peripheral blood CAR T-cell expansion during the first 30 days post-infusion is illustrated in Figure S2.</p><p>All patients developed cytokine release syndrome (Grade 1 in three patients, Grade 2 in four patients). ICANS of any grade occurred in six patients (Grade 3 in one, Grade 4 in two). The Grade 4 ICANS cases involved a 65-year-old patient with a history of cerebral involvement and whole-brain radiotherapy who received axi-cel for isolated ocular relapse, and a 71-year-old patient who received axi-cel with progressive bulky cerebral disease at the time of infusion. Four out of seven patients received steroids, and no patients developed ocular pseudoprogression.</p><p>This is the first series on CAR T-cell therapy in patients with VRL, and we acknowledge the small and heterogeneous series (VRL both primary and secondary, with or without CNS involvement), which may affect the generalizability of our findings. In particular, it is conceivable that the presence of occult systemic disease, especially in cases of previous systemic involvement, may have facilitated CAR T-cell expansion or that distinct molecular profiles may have influenced the duration of the therapeutic response.</p><p>To date, only a few case reports on CAR T-cell therapy for hematological malignancies with ocular involvement have been published.<span><sup>11-13</sup></span> Taher et al. reported a case of early ocular failure of CAR T-cells in a patient with diffuse large B-cell lymphoma (DLBCL) treated for intraocular and CNS localizations, where the CNS disease remained in CR.<span><sup>14</sup></span> Additionally, two cases of CAR T-cell therapy for B-cell acute lymphoblastic leukemia with ophthalmic localization have been described: one demonstrated persistent CR after 1 year, whereas the other reported a flare reaction with sudden ocular deterioration following CAR T-cell therapy.<span><sup>12, 13</sup></span> The flare reaction consisted of bilateral retinal detachment occurring on Day 7 after CAR T-cell infusion, in the absence of blasts in the AH, but with the presence of white blood cells, 35% of which expressing CD3.</p><p>Although our patients were highly pretreated and refractory, the efficacy of CAR T-cells appeared very promising, with outcomes comparable to those achieved with ASCT within our network (2-year PFS and OS rates of 68% and 87%, respectively).<span><sup>15</sup></span> These results also compared favorably to those obtained in the first-line treatment of primary vitreoretinal lymphoma.<span><sup>1</sup></span> The impact of bridging therapy and the remission status at the time of CAR T-cell infusion may represent confounding factors, but the persistence of response after CAR T-cell infusion in the three highly refractory patients with PR before CAR T-cells (23, 16, and 17 months, respectively) strongly supports the major contribution of CAR T-cells in maintaining disease control. Notably, one patient in CR at 6 months exhibited high IL-10 in the AH (1246 pg/mL) without any clinical signs of relapse but subsequently relapsed at 10 months. The IPCG criteria for ophthalmic response do not account for IL-10 quantification; however, the relapse might have already been present in an infra-clinical state as early as 6 months.</p><p>AH CAR T-cell trafficking was demonstrated for the first time in this series in one patient evaluated at multiple time points, confirming their ability to migrate and persist in the AH despite low blood levels of the target antigen. We can hypothesize that CAR T-cells were detected in the AH of only one of three tested patients due to several factors, including the exceptional localizations in the AH of VRL, technical challenges associated with analyzing small-volume samples, sensitivity of flow cytometry, and potentially suboptimal sampling timing relative to the kinetics of CAR T-cell expansion. Further investigations are needed to better characterize CAR T-cell trafficking in these scenarios.</p><p>Finally, we noted unusual Grade 3–4 ICANS in th1ree of seven patients, while the rate and severity of neurotoxicity reported in CNS lymphomas are comparable to those in systemic lymphomas.<span><sup>5, 7, 16</sup></span> This unexpectedly high rate of severe ICANS in our cohort may be attributed to multiple factors, including the small sample size, a high number of prior treatment lines, a history of ASCT and/or encephalic radiotherapy (both known to be associated with cognitive impairment), the presence of CNS disease (reflecting tumor burden), and the type of CAR T-cell construct used. Notably, among the three patients who developed Grade 3-4 ICANS in our cohort, all had progressive CNS disease; two had previously received ASCT with or without encephalic radiotherapy and were treated with axi-cel. Conversely, we did note any ocular immune-related adverse events during follow-up.</p><p>Larger studies are needed to evaluate the true risk of severe neurotoxicity following CAR T-cell therapy in VRLs. Nevertheless, the decision to pursue CAR T-cell therapy should carefully consider this risk.</p><p>In conclusion, this study is the first to report a significant response rate after CAR T-cell therapy in highly pretreated and refractory VRLs, with a promising 2-year PFS rate of 69%. Longer follow-up is essential to confirm these findings and define the role of CAR T-cell therapy within the broader therapeutic strategy for VRL.</p><p><b>Cécile Pivert</b>: Investigation; formal analysis; writing—original draft; software. <b>Adélaïde Toutée</b>: Investigation; data curation. <b>Christophe Parizot</b>: Formal analysis; investigation; data curation. <b>Denis Malaise</b>: Investigation. <b>Alexandre Matet</b>: Investigation. <b>Valérie Touitou</b>: Investigation. <b>Magali Le Garff-Tavernier</b>: Formal analysis; data curation. <b>Damien Roos-Weil</b>: Investigation. <b>Sarah Touhami</b>: Investigation. <b>Nabih Azar</b>: Investigation. <b>Ines Boussen</b>: Investigation. <b>Véronique Morel</b>: Investigation. <b>Madalina Uzunov</b>: Investigation. <b>Clotilde Bravetti</b>: Formal analysis; data curation. <b>Carole Metz</b>: Investigation. <b>Eve Todesco</b>: Formal analysis; data curation. <b>Delphine Sterlin</b>: Formal analysis. <b>Carole Soussain</b>: Investigation. <b>Kahina Ouis Tarhi</b>: Investigation. <b>Anne Besançon</b>: Investigation. <b>Khê Hoang-Xuan</b>: Investigation. <b>Sylvain Choquet</b>: Conceptualization; supervision; writing—review and editing. <b>Caroline Houillier</b>: Conceptualization; supervision; writing—review and editing; formal analysis; validation; methodology; software. <b>Marine Baron</b>: Conceptualization; validation; supervision; writing—review and editing; methodology; software.</p><p>The authors declare no conflicts of interest.</p><p>No funding.</p>\",\"PeriodicalId\":12982,\"journal\":{\"name\":\"HemaSphere\",\"volume\":\"9 8\",\"pages\":\"\"},\"PeriodicalIF\":14.6000,\"publicationDate\":\"2025-08-25\",\"publicationTypes\":\"Journal Article\",\"fieldsOfStudy\":null,\"isOpenAccess\":false,\"openAccessPdf\":\"https://onlinelibrary.wiley.com/doi/epdf/10.1002/hem3.70208\",\"citationCount\":\"0\",\"resultStr\":null,\"platform\":\"Semanticscholar\",\"paperid\":null,\"PeriodicalName\":\"HemaSphere\",\"FirstCategoryId\":\"3\",\"ListUrlMain\":\"https://onlinelibrary.wiley.com/doi/10.1002/hem3.70208\",\"RegionNum\":2,\"RegionCategory\":\"医学\",\"ArticlePicture\":[],\"TitleCN\":null,\"AbstractTextCN\":null,\"PMCID\":null,\"EPubDate\":\"\",\"PubModel\":\"\",\"JCR\":\"Q1\",\"JCRName\":\"HEMATOLOGY\",\"Score\":null,\"Total\":0}","platform":"Semanticscholar","paperid":null,"PeriodicalName":"HemaSphere","FirstCategoryId":"3","ListUrlMain":"https://onlinelibrary.wiley.com/doi/10.1002/hem3.70208","RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q1","JCRName":"HEMATOLOGY","Score":null,"Total":0}
引用次数: 0

摘要

玻璃体视网膜淋巴瘤(VRL)是一种罕见的大b细胞淋巴瘤(LBCL)亚型,可表现为孤立状态或中枢神经系统(CNS)或全身受累。与大脑一样,眼睛也是一个免疫特权部位,根据血眼屏障提出了独特的治疗挑战。尽管其病程通常为惰性,但VRL难以根治,由于累及中枢神经系统的风险高,其长期预后较差原发性VRL的中位总生存期(OS)为36-75个月,原发性中枢神经系统淋巴瘤孤立性玻璃体视网膜复发的中位总生存期约为57个月因此,迫切需要新的治疗策略。抗cd19嵌合抗原受体(CAR - t细胞)在治疗系统性lbcl方面取得了重大进展。研究表明,CAR - t细胞可以穿过血脑屏障,在中枢神经系统淋巴瘤中表现出高效率,5-8但它们在VRL治疗中的潜力尚不完全清楚,而且它们的效率从未从眼科的角度用最新的验证检查来描述。自2020年以来,CAR - t细胞疗法已在法国眼脑淋巴瘤网络(LOC网络)中被考虑用于复发或难治性VRL。本研究回顾性分析了CAR - t细胞治疗原发性或继发性VRL的患者。我们通过LOC网络数据库回顾性地确定了截至2023年11月在Pitié-Salpêtrière医院(巴黎)接受商业化抗cd19 CAR - t细胞治疗的所有VRL患者。符合条件的患者有活跃的玻璃体视网膜受累,无论是孤立的还是伴有中枢神经系统受累。VRL可能是原发性或继发性系统性或中枢神经系统淋巴瘤。回顾性收集2024年4月至10月期间的医疗记录数据。LOC数据库已得到协调中心机构伦理委员会和法国“全国信息和自由自由委员会”的核可。所有患者均提供书面知情同意书。采用国际原发性中枢神经系统淋巴瘤协作组(IPCG)标准对患者的反应进行评估,并且在CAR - t细胞治疗前后立即进行的所有眼科检查均由piti<s:1> - salpsamtri<e:1>医院的一名眼科医生根据每次会诊时拍摄的照片进行系统审查(补充方法)。无进展生存期(PFS)、眼部无进展生存期(OFS)和OS被定义为CAR - t细胞输注的时间:“分别到中枢神经系统、眼部或全身性疾病进展或复发/到眼部进展或复发/和死亡。”生存率采用Kaplan-Meier法计算。使用GraphPad Prism v10.0进行统计分析。细胞因子释放综合征和免疫效应细胞相关神经毒性综合征(ICANS)根据2019年美国移植和细胞治疗学会(ASTCT)指南进行分级。在FACSCanto II型细胞仪上,使用灵敏的流式细胞仪头阵列试剂盒(BD Biosciences)对房水(AH)进行白细胞介素(IL)-10和IL-6检测。根据实验室标准阈值将AH IL-10水平分为可检测(≥2.5 pg/mL)或不可检测(&lt;2.5 pg/mL)。用流式细胞术检测血液和AH中CAR - t细胞的存在。每个样品用1µL CD19 CAR检测试剂(Miltenyi Biotec)孵育15分钟,然后用以下抗体染色:抗生物素- pe (Miltenyi Biotec)、CD45-KrO(克隆J33)、CD3-Alexa Fluor A750(克隆UCHT1)、CD4-ECD(克隆SFCI12T4D11)和CD8-Alexa Fluor 700(克隆B9.11)。最终样品在DxFLEX流式细胞仪(Beckman Coulter)上分析,使用CytExpert (Beckman Coulter)软件版本2.0.2.18处理数据。在2020年8月至2023年12月期间,7名VRL患者接受了CAR - t细胞治疗。表1和表S1总结了他们的主要基线特征和个体结局。在CAR - t细胞治疗决定时,4名患者有分离性VRL,而3名患者也有脑实质受损伤。他们的中位年龄为70岁(范围50-76),东部肿瘤合作组的中位成绩为1(范围1 - 3)。白血病穿刺前接受治疗的中位数为4(范围2-6),包括4例患者的自体干细胞移植(ASCT)(基于硫替帕的诱导方案)(2例患者在CAR - t细胞治疗时不符合ASCT的条件)。除一名患者外,所有患者均接受了针对眼/脑疾病的药物治疗,如甲氨蝶呤、替莫唑胺、依鲁替尼或细胞毒性化疗(异环磷酰胺、依托泊苷和卡铂)(表1)。没有类固醇治疗方案。在CAR - t细胞输注时,对于眼部受累,2例完全缓解(CR), 2例部分缓解(PR), 2例病情稳定,1例病情进展。 此外,3例患者有中枢神经系统进行性疾病。3例患者接受tisagenlecleucel治疗,4例患者接受axicabtagene ciloleucel(轴细胞)治疗。在1个月时,所有5例可评估患者均观察到眼科反应,包括2例PR(40%)和3例CR(60%)。2例出现严重神经毒性且此时无法进行眼科评估的患者数据不可用(表S1)。在随访期间,所有7例患者的最佳缓解是CR或未确诊的CR。从CAR - t细胞输注到首次反应的中位时间为1个月(范围1 - 4)。中位随访23个月(10-47个月)后,2年的OFS、PFS和OS率分别为83%、69%和83%(图1A、B)。在car - t细胞治疗后的前6个月,6名受测患者的AH中的IL-10剂量检测不到,只有一名患者在几个月后复发(图S1)。另一方面,在此期间检测的所有6例患者的AH中均检测到IL-6水平超过2.5 pg/mL(范围19-194 pg/mL),与≥3级ICANS的发生没有观察到相关性。7例患者中有2例出现复发:1例患者在CAR - t细胞输注10个月后出现孤立性眼内复发,另1例患者在CAR - t细胞输注5个月后出现脑和脑脊液复发。为了更好地表征CAR - t细胞在眼睛中的运输和持久性,在三名患者的不同时间点进行了流式细胞术(一名患者进行了四次评估,两名患者进行了两次评估)。1例患者在11天、1个月、4个月和10个月(最后一次眼科随访)时显示AH CAR - t细胞阳性(图1C)。另外两名患者在所有时间点的CAR - t细胞检测结果均为阴性。在最后一次眼科随访时(CAR - t细胞输注后分别为17、16和10个月),所有3例患者均保持CR。外周血CAR - t细胞在输注后30天内的扩增情况如图S2所示。所有患者均出现细胞因子释放综合征(3例为1级,4例为2级)。6例患者发生了任何级别的ICANS(1例3级,2例4级)。4级ICANS病例包括一名65岁有脑受累和全脑放疗史的患者,因孤立性眼部复发接受轴细胞治疗,以及一名71岁接受轴细胞治疗的患者,在输注时伴有进行性大体积脑疾病。7名患者中有4名接受了类固醇治疗,没有患者出现眼部假性进展。这是CAR - t细胞治疗VRL患者的第一个系列,我们承认这个小而异质性的系列(原发性和继发性VRL,有无中枢神经系统受累)可能会影响我们研究结果的普遍性。特别是,可以想象的是,隐蔽性全身性疾病的存在,特别是在先前的全身性疾病的情况下,可能促进了CAR - t细胞的扩增,或者不同的分子谱可能影响了治疗反应的持续时间。迄今为止,只有少数病例报告CAR - t细胞治疗血液病恶性肿瘤累及眼部已发表。11-13 Taher等人报道了一例弥漫性大b细胞淋巴瘤(DLBCL)患者在眼内和中枢神经系统定位治疗中出现CAR - t细胞早期眼衰竭的病例,其中中枢神经系统疾病仍在cr中。14此外,还报道了两例CAR - t细胞治疗伴有眼部定位的b细胞急性淋巴细胞白血病的病例:一名患者在1年后表现出持续性CR,而另一名患者在CAR - t细胞治疗后出现突发性视力恶化的急性反应。12,13耀斑反应包括在CAR - t细胞输注后第7天发生的双侧视网膜脱离,AH中没有母细胞,但存在白细胞,其中35%表达CD3。尽管我们的患者经过了高度预处理且难治性,但CAR - t细胞的疗效似乎非常有希望,其结果与我们网络中的ASCT相当(2年PFS和OS率分别为68%和87%)这些结果也与原发性玻璃体视网膜淋巴瘤的一线治疗结果相比较桥接治疗的影响和CAR - t细胞输注时的缓解状态可能是混淆因素,但在3例高度难治性PR患者中,CAR - t细胞输注后的持续反应(分别为23、16和17个月)有力地支持了CAR - t细胞在维持疾病控制方面的主要贡献。值得注意的是,一名CR患者在6个月时表现出AH中高IL-10 (1246 pg/mL),没有任何临床复发迹象,但随后在10个月时复发。 IPCG眼科反应标准没有考虑到IL-10的量化;然而,复发可能早在6个月就已经出现在临床外状态。在本系列研究中,CAR - t细胞运输首次在一名患者中得到证实,该患者在多个时间点进行了评估,证实了它们在血液中低水平靶抗原的情况下迁移和持续存在AH的能力。我们可以假设,由于几个因素,包括VRL的AH异常定位,与分析小体积样品相关的技术挑战,流式细胞术的敏感性,以及相对于CAR - t细胞扩增动力学的潜在次优采样时间,在三名测试患者中仅检测到CAR - t细胞。在这些情况下,需要进一步的调查来更好地表征CAR - t细胞运输。最后,我们注意到7例患者中有3例出现不寻常的3-4级ICANS,而中枢神经系统淋巴瘤的神经毒性发生率和严重程度与系统性淋巴瘤相当。5,7,16在我们的队列中,严重ICANS的意外高发生率可能归因于多种因素,包括小样本量、大量的先前治疗线、ASCT和/或脑放射治疗史(已知与认知障碍有关)、中枢神经系统疾病的存在(反映肿瘤负担)以及所使用的CAR - t细胞结构类型。值得注意的是,在我们队列中发生3-4级ICANS的3例患者中,所有患者都患有进行性中枢神经系统疾病;其中2例曾接受ASCT伴或不伴脑放射治疗,并接受轴细胞治疗。相反,我们在随访期间确实注意到任何眼部免疫相关的不良事件。需要更大规模的研究来评估CAR - t细胞治疗vrl后严重神经毒性的真实风险。然而,在决定采用CAR - t细胞疗法时应该仔细考虑这种风险。总之,这项研究首次报道了CAR - t细胞治疗高度预处理和难治性vrl后的显着应答率,2年PFS率为69%。为了证实这些发现和确定CAR - t细胞疗法在更广泛的治疗策略中的作用,更长的随访是必不可少的。正式的分析;原创作品草案;软件。Adélaïde tout<s:2>:调查;数据管理。Christophe Parizot:形式分析;调查;数据管理。丹尼斯:调查。Alexandre Matet:调查。valsamrie Touitou:调查。Magali Le Garff-Tavernier:形式分析;数据管理。达米恩·鲁斯-威尔:调查。Sarah Touhami:调查。拿比哈:调查。伊内斯·布森:调查。vsamronique Morel:调查。Madalina Uzunov:调查。Clotilde Bravetti:形式分析;数据管理。卡罗尔·梅斯:调查。Eve Todesco:形式分析;数据管理。德尔芬·斯特林:形式分析。卡罗尔·苏塞恩:调查。Kahina louis Tarhi:调查。安妮:调查。Khê黄轩:调查。Sylvain Choquet:概念化;监督;写作-审查和编辑。Caroline Houillier:概念化;监督;写作——审阅和编辑;正式的分析;验证;方法;软件。海洋男爵:概念化;验证;监督;写作——审阅和编辑;方法;软件。作者声明无利益冲突。没有资金。
本文章由计算机程序翻译,如有差异,请以英文原文为准。

Preliminary evidence of CAR T-cell therapy activity in vitreoretinal lymphomas: An LOC network study

Preliminary evidence of CAR T-cell therapy activity in vitreoretinal lymphomas: An LOC network study

Vitreoretinal lymphoma (VRL) is a rare subtype of large B-cell lymphoma (LBCL) that may present as an isolated condition or with central nervous system (CNS) or systemic involvement. Like the brain, the eye is an immune-privileged site, posing unique therapeutic challenges according to the blood–eye barrier. Despite its typically indolent course, VRL is difficult to eradicate, and the long-term prognosis is poor because of the high risk of CNS involvement.1 The median overall survival (OS) is 36–75 months for primary VRL and approximately 57 months for isolated vitreoretinal relapses of primary CNS lymphomas.2 Therefore, new therapeutic strategies are urgently needed.

Anti-CD19 chimeric antigen receptor (CAR) T-cells represent a major advancement in managing systemic LBCL.3, 4 Studies show that CAR T-cells can cross the blood–brain barrier and demonstrate high efficacy in CNS lymphomas,5-8 but their potential in VRL treatment is not fully understood, and their efficiency has never been described from an ophthalmologic point of view with the newest validated exams.

Since 2020, CAR T-cell therapy has been considered within the French oculocerebral lymphoma network (LOC network) for relapsed or refractory VRL. This study retrospectively analyzed patients with primary or secondary VRL treated with CAR T-cells.

We retrospectively identified all VRL patients treated with commercial anti-CD19 CAR T-cells at Pitié-Salpêtrière Hospital (Paris) via the LOC network database until November 2023. Eligible patients had active vitreoretinal involvement, either isolated or with CNS involvement. VRL could be primary or secondary to systemic or CNS lymphoma. Data were retrospectively collected from medical records between April and October 2024. The LOC database was approved by the Institutional Ethical Committee of the coordinating center and the French “Commission Nationale de l'Informatique et des Libertés” (CNIL). All patients provided written informed consent. Responses were evaluated using International Primary CNS Lymphoma Collaborative Group (IPCG) criteria,9 and all ophthalmologic examinations of the patient performed immediately before and after the CAR T-cells were systematically reviewed by a single ophthalmologist from the Pitié-Salpétrière Hospital, based on the photographs taken at each consultation (Supplementary Methods). Progression-free survival (PFS), ophthalmic-free survival (OFS), and OS were defined as the time from CAR T-cell infusion: “to CNS, ophthalmic or systemic disease progression or relapse/to ophthalmic progression or relapse/and to death respectively.” Survival rates were calculated using the Kaplan–Meier method. Statistical analyses were conducted using GraphPad Prism v10.0. Cytokine release syndrome and immune effector cell-associated neurotoxicity syndrome (ICANS) were graded per 2019 American Society for Transplantation and Cellular Therapy (ASTCT) guidelines.10

Interleukin (IL)-10 and IL-6 assays were performed on the aqueous humor (AH) using a sensitive Cytometric Bead Array kit (BD Biosciences) on a FACSCanto II cytometer. The AH IL-10 level was classified as detectable (≥2.5 pg/mL) or undetectable (<2.5 pg/mL) based on the laboratory's standard threshold. The presence of CAR T-cells in the blood and AH was measured by flow cytometry. Each sample was incubated for 15 min with 1 µL of CD19 CAR Detection Reagent (Miltenyi Biotec), followed by staining with the following antibodies: anti-Biotin-PE (Miltenyi Biotec), CD45-KrO (clone J33), CD3-Alexa Fluor A750 (clone UCHT1), CD4-ECD (clone SFCI12T4D11), and CD8-Alexa Fluor 700 (clone B9.11). The final sample was analyzed on a DxFLEX flow cytometer (Beckman Coulter), and data were processed using CytExpert (Beckman Coulter) software version 2.0.2.18.

Between August 2020 and December 2023, seven patients with VRL were treated with CAR T-cells. Their main baseline characteristics and individual outcomes are summarized in Tables 1 and S1.

At the time of the CAR T-cell therapy decision, four patients had isolated VRL, whereas three also had brain parenchymal involvement. Their median age was 70 years (range, 50–76), and their median Eastern Cooperative Oncology Group performance status was 1 (range, 1–3). The median number of prior therapies before leukapheresis was 4 (range, 2–6), including autologous stem cell transplantation (ASCT) (thiotepa-based induction regimen) in four patients (two patients were ineligible for ASCT at the time of CAR T-cells). All but one patient received bridging therapy using drug regimens directed against ocular/brain disease such as methotrexate, temozolomide, ibrutinib, or cytotoxic chemotherapies (ifosfamide, etoposide, and carboplatin) (Table 1). There were no steroid-containing regimens.

At the time of CAR T-cell infusion, with regard to ophthalmic involvement, two patients had a complete response (CR), two had a partial response (PR), two had stable disease, and one had progressive disease. Additionally, three patients had CNS progressive disease.

Three patients received tisagenlecleucel and four received axicabtagene ciloleucel (axi-cel). At 1 month, an ophthalmologic response was observed in all five evaluable patients, including PR in two (40%) and CR in three (60%). Data were unavailable for two patients who developed severe neurotoxicity and could not undergo ophthalmologic assessment at this time (Table S1). During follow-up, the best response achieved was CR or CR unconfirmed for all seven patients. The median time from CAR T-cell infusion to first response was 1 month (range, 1–4). After a median follow-up of 23 months (range, 10–47), the 2-year OFS, PFS, and OS rates were 83%, 69% and 83%, respectively (Figure 1A,B). IL-10 dosage in the AH in six tested patients during the first 6 months post-CAR T-cell therapy was undetectable for all but one patient who relapsed a few months later (Figure S1). On the other hand, IL-6 levels exceeding 2.5 pg/mL were detected in the AH of all six patients tested during this period (range, 19–194 pg/mL), with no observed correlation to the occurrence of grade ≥ 3 ICANS. Two of seven patients experienced relapses: one patient had an isolated intraocular relapse 10 months after CAR T-cell infusion, and the other one experienced a relapse in the brain and cerebrospinal fluid 5 months after CAR T-cell infusion.

To better characterize the trafficking and persistence of CAR T-cells in the eye, flow cytometry was performed at different time points in three patients (one patient was assessed four times, and two were assessed twice). One patient demonstrated AH positivity for CAR T-cells at 11 days, 1 month, 4 months, and 10 months (last ophthalmologic follow-up) (Figure 1C). The other two patients tested negative for CAR T-cells at all time points. All three patients remained in CR at their last ophthalmologic follow-up (17, 16, and 10 months after CAR T-cell infusion, respectively). Peripheral blood CAR T-cell expansion during the first 30 days post-infusion is illustrated in Figure S2.

All patients developed cytokine release syndrome (Grade 1 in three patients, Grade 2 in four patients). ICANS of any grade occurred in six patients (Grade 3 in one, Grade 4 in two). The Grade 4 ICANS cases involved a 65-year-old patient with a history of cerebral involvement and whole-brain radiotherapy who received axi-cel for isolated ocular relapse, and a 71-year-old patient who received axi-cel with progressive bulky cerebral disease at the time of infusion. Four out of seven patients received steroids, and no patients developed ocular pseudoprogression.

This is the first series on CAR T-cell therapy in patients with VRL, and we acknowledge the small and heterogeneous series (VRL both primary and secondary, with or without CNS involvement), which may affect the generalizability of our findings. In particular, it is conceivable that the presence of occult systemic disease, especially in cases of previous systemic involvement, may have facilitated CAR T-cell expansion or that distinct molecular profiles may have influenced the duration of the therapeutic response.

To date, only a few case reports on CAR T-cell therapy for hematological malignancies with ocular involvement have been published.11-13 Taher et al. reported a case of early ocular failure of CAR T-cells in a patient with diffuse large B-cell lymphoma (DLBCL) treated for intraocular and CNS localizations, where the CNS disease remained in CR.14 Additionally, two cases of CAR T-cell therapy for B-cell acute lymphoblastic leukemia with ophthalmic localization have been described: one demonstrated persistent CR after 1 year, whereas the other reported a flare reaction with sudden ocular deterioration following CAR T-cell therapy.12, 13 The flare reaction consisted of bilateral retinal detachment occurring on Day 7 after CAR T-cell infusion, in the absence of blasts in the AH, but with the presence of white blood cells, 35% of which expressing CD3.

Although our patients were highly pretreated and refractory, the efficacy of CAR T-cells appeared very promising, with outcomes comparable to those achieved with ASCT within our network (2-year PFS and OS rates of 68% and 87%, respectively).15 These results also compared favorably to those obtained in the first-line treatment of primary vitreoretinal lymphoma.1 The impact of bridging therapy and the remission status at the time of CAR T-cell infusion may represent confounding factors, but the persistence of response after CAR T-cell infusion in the three highly refractory patients with PR before CAR T-cells (23, 16, and 17 months, respectively) strongly supports the major contribution of CAR T-cells in maintaining disease control. Notably, one patient in CR at 6 months exhibited high IL-10 in the AH (1246 pg/mL) without any clinical signs of relapse but subsequently relapsed at 10 months. The IPCG criteria for ophthalmic response do not account for IL-10 quantification; however, the relapse might have already been present in an infra-clinical state as early as 6 months.

AH CAR T-cell trafficking was demonstrated for the first time in this series in one patient evaluated at multiple time points, confirming their ability to migrate and persist in the AH despite low blood levels of the target antigen. We can hypothesize that CAR T-cells were detected in the AH of only one of three tested patients due to several factors, including the exceptional localizations in the AH of VRL, technical challenges associated with analyzing small-volume samples, sensitivity of flow cytometry, and potentially suboptimal sampling timing relative to the kinetics of CAR T-cell expansion. Further investigations are needed to better characterize CAR T-cell trafficking in these scenarios.

Finally, we noted unusual Grade 3–4 ICANS in th1ree of seven patients, while the rate and severity of neurotoxicity reported in CNS lymphomas are comparable to those in systemic lymphomas.5, 7, 16 This unexpectedly high rate of severe ICANS in our cohort may be attributed to multiple factors, including the small sample size, a high number of prior treatment lines, a history of ASCT and/or encephalic radiotherapy (both known to be associated with cognitive impairment), the presence of CNS disease (reflecting tumor burden), and the type of CAR T-cell construct used. Notably, among the three patients who developed Grade 3-4 ICANS in our cohort, all had progressive CNS disease; two had previously received ASCT with or without encephalic radiotherapy and were treated with axi-cel. Conversely, we did note any ocular immune-related adverse events during follow-up.

Larger studies are needed to evaluate the true risk of severe neurotoxicity following CAR T-cell therapy in VRLs. Nevertheless, the decision to pursue CAR T-cell therapy should carefully consider this risk.

In conclusion, this study is the first to report a significant response rate after CAR T-cell therapy in highly pretreated and refractory VRLs, with a promising 2-year PFS rate of 69%. Longer follow-up is essential to confirm these findings and define the role of CAR T-cell therapy within the broader therapeutic strategy for VRL.

Cécile Pivert: Investigation; formal analysis; writing—original draft; software. Adélaïde Toutée: Investigation; data curation. Christophe Parizot: Formal analysis; investigation; data curation. Denis Malaise: Investigation. Alexandre Matet: Investigation. Valérie Touitou: Investigation. Magali Le Garff-Tavernier: Formal analysis; data curation. Damien Roos-Weil: Investigation. Sarah Touhami: Investigation. Nabih Azar: Investigation. Ines Boussen: Investigation. Véronique Morel: Investigation. Madalina Uzunov: Investigation. Clotilde Bravetti: Formal analysis; data curation. Carole Metz: Investigation. Eve Todesco: Formal analysis; data curation. Delphine Sterlin: Formal analysis. Carole Soussain: Investigation. Kahina Ouis Tarhi: Investigation. Anne Besançon: Investigation. Khê Hoang-Xuan: Investigation. Sylvain Choquet: Conceptualization; supervision; writing—review and editing. Caroline Houillier: Conceptualization; supervision; writing—review and editing; formal analysis; validation; methodology; software. Marine Baron: Conceptualization; validation; supervision; writing—review and editing; methodology; software.

The authors declare no conflicts of interest.

No funding.

求助全文
通过发布文献求助,成功后即可免费获取论文全文。 去求助
来源期刊
HemaSphere
HemaSphere Medicine-Hematology
CiteScore
6.10
自引率
4.50%
发文量
2776
审稿时长
7 weeks
期刊介绍: HemaSphere, as a publication, is dedicated to disseminating the outcomes of profoundly pertinent basic, translational, and clinical research endeavors within the field of hematology. The journal actively seeks robust studies that unveil novel discoveries with significant ramifications for hematology. In addition to original research, HemaSphere features review articles and guideline articles that furnish lucid synopses and discussions of emerging developments, along with recommendations for patient care. Positioned as the foremost resource in hematology, HemaSphere augments its offerings with specialized sections like HemaTopics and HemaPolicy. These segments engender insightful dialogues covering a spectrum of hematology-related topics, including digestible summaries of pivotal articles, updates on new therapies, deliberations on European policy matters, and other noteworthy news items within the field. Steering the course of HemaSphere are Editor in Chief Jan Cools and Deputy Editor in Chief Claire Harrison, alongside the guidance of an esteemed Editorial Board comprising international luminaries in both research and clinical realms, each representing diverse areas of hematologic expertise.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
确定
请完成安全验证×
copy
已复制链接
快去分享给好友吧!
我知道了
右上角分享
点击右上角分享
0
联系我们:info@booksci.cn Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。 Copyright © 2023 布克学术 All rights reserved.
京ICP备2023020795号-1
ghs 京公网安备 11010802042870号
Book学术文献互助
Book学术文献互助群
群 号:604180095
Book学术官方微信