Lenalidomide is an oral immunomodulatory agent approved for the treatment of relapsed/refractory adult T-cell leukemia/lymphoma (ATLL) in Japan. Post-marketing surveillance (PMS) was conducted to confirm its safety and effectiveness. From April 2017 until April 2020, safety data were obtained for 77 patients and effectiveness data for 65 patients (31.2% of patients had progressive disease as the best response to their most recent prior regimen). Forty-nine patients (63.6%) in the safety analysis set experienced an adverse drug reaction (ADR). Grade ≥ 3 ADRs occurred in 42.9%. The most common Grade ≥ 3 ADRs were neutrophil count decreased/neutropenia and platelet count decreased/thrombocytopenia (11.7% each). Serious ADRs occurred in 26 patients. Five patients had previously received allogeneic hematopoietic stem cell transplantation. Among these, one experienced acute graft versus host disease (GvHD) during lenalidomide administration and two responded to lenalidomide. Effectiveness analysis showed that an objective response was achieved in 29.2% of patients. No statistically significant differences were observed in the objective response rates of patients aged < 70 versus those aged ≥ 70 years (33.3% vs 28.0%, respectively; p = 0.6904). No new safety signals were observed in this PMS, and lenalidomide demonstrated a favorable benefit-risk balance in Japanese patients with ATLL.
{"title":"Safety and effectiveness of lenalidomide in Japanese patients with relapsed/refractory ATLL: post-marketing surveillance.","authors":"Tohru Miyazaki, Shuji Uno, Hiroaki Fujimori, Yoko Motegi","doi":"10.1007/s12185-024-03864-3","DOIUrl":"https://doi.org/10.1007/s12185-024-03864-3","url":null,"abstract":"<p><p>Lenalidomide is an oral immunomodulatory agent approved for the treatment of relapsed/refractory adult T-cell leukemia/lymphoma (ATLL) in Japan. Post-marketing surveillance (PMS) was conducted to confirm its safety and effectiveness. From April 2017 until April 2020, safety data were obtained for 77 patients and effectiveness data for 65 patients (31.2% of patients had progressive disease as the best response to their most recent prior regimen). Forty-nine patients (63.6%) in the safety analysis set experienced an adverse drug reaction (ADR). Grade ≥ 3 ADRs occurred in 42.9%. The most common Grade ≥ 3 ADRs were neutrophil count decreased/neutropenia and platelet count decreased/thrombocytopenia (11.7% each). Serious ADRs occurred in 26 patients. Five patients had previously received allogeneic hematopoietic stem cell transplantation. Among these, one experienced acute graft versus host disease (GvHD) during lenalidomide administration and two responded to lenalidomide. Effectiveness analysis showed that an objective response was achieved in 29.2% of patients. No statistically significant differences were observed in the objective response rates of patients aged < 70 versus those aged ≥ 70 years (33.3% vs 28.0%, respectively; p = 0.6904). No new safety signals were observed in this PMS, and lenalidomide demonstrated a favorable benefit-risk balance in Japanese patients with ATLL.</p>","PeriodicalId":13992,"journal":{"name":"International Journal of Hematology","volume":" ","pages":""},"PeriodicalIF":1.7,"publicationDate":"2024-11-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142681771","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Although tyrosine kinase inhibitors (TKIs) have improved the prognosis of chronic myeloid leukemia (CML), some patients do not respond to TKIs. We evaluated 21 patients with CML treated with asciminib, which is effective in heavily pretreated patients. The median age was 63 years (range 25-82). Fourteen patients (67%) had been treated with at least three TKIs (range 2-5 prior lines). The switch to asciminib was due to intolerance in 14 patients (67%) and failure in seven patients (33%). The median duration of asciminib exposure was 237 days. With a median follow-up of 273 days, three patients (14%) discontinued asciminib due to failure and two (10%) due to intolerance. Among the 20 evaluable patients, the cumulative rates of molecular response with a two-log reduction, major molecular response, and four-log reduction were 80%, 60%, and 15%, respectively. The six-month event-free survival rate was 74.7%. The most frequent adverse events were liver dysfunction (29%), elevated amylase levels (14%), and renal dysfunction (10%). No patient experienced cardiovascular events. Six patients (29%) experienced cross-intolerance to asciminib, a rate similar to that for previous TKIs. Our study supports the efficacy and tolerability of asciminib in heavily pretreated CML patients in real-world settings.
{"title":"Real-world toxicity and efficacy of asciminib in heavily pretreated patients with chronic myeloid leukemia.","authors":"Yoshimi Ishii, Shin Fujisawa, Takuya Miyazaki, Yuki Nakajima, Ayako Matsumura, Katsumichi Fujimaki, Taisei Suzuki, Maki Hagihara, Marika Tanaka, Chizuko Hashimoto, Hideaki Nakajima","doi":"10.1007/s12185-024-03873-2","DOIUrl":"10.1007/s12185-024-03873-2","url":null,"abstract":"<p><p>Although tyrosine kinase inhibitors (TKIs) have improved the prognosis of chronic myeloid leukemia (CML), some patients do not respond to TKIs. We evaluated 21 patients with CML treated with asciminib, which is effective in heavily pretreated patients. The median age was 63 years (range 25-82). Fourteen patients (67%) had been treated with at least three TKIs (range 2-5 prior lines). The switch to asciminib was due to intolerance in 14 patients (67%) and failure in seven patients (33%). The median duration of asciminib exposure was 237 days. With a median follow-up of 273 days, three patients (14%) discontinued asciminib due to failure and two (10%) due to intolerance. Among the 20 evaluable patients, the cumulative rates of molecular response with a two-log reduction, major molecular response, and four-log reduction were 80%, 60%, and 15%, respectively. The six-month event-free survival rate was 74.7%. The most frequent adverse events were liver dysfunction (29%), elevated amylase levels (14%), and renal dysfunction (10%). No patient experienced cardiovascular events. Six patients (29%) experienced cross-intolerance to asciminib, a rate similar to that for previous TKIs. Our study supports the efficacy and tolerability of asciminib in heavily pretreated CML patients in real-world settings.</p>","PeriodicalId":13992,"journal":{"name":"International Journal of Hematology","volume":" ","pages":""},"PeriodicalIF":1.7,"publicationDate":"2024-11-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142667918","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Ganciclovir and foscarnet are two representative anti-cytomegalovirus (CMV) agents. A previous regional study revealed a lower risk of chronic graft-versus-host disease (GVHD) in patients who received pre-emptive foscarnet. We conducted a retrospective nationwide study to confirm the results. A total of 8890 patients aged 16 or older with hematological malignancies who received foscarnet (n = 1555) or ganciclovir (n = 7335) during their first hematopoietic stem cell transplantation (HSCT) were included. The risks of chronic GVHD (hazard ratio [HR], 1.26; 95% confidence interval [CI], 1.13-1.40; P < 0.001) and extensive chronic GVHD (HR, 1.16; 95% CI, 1.01-1.33; P = 0.033) were higher with ganciclovir. Among male patients with a female donor, the incidence of extensive chronic GVHD 3 years after HSCT was clearly lower with foscarnet (13%; 95% CI, 9-16%) than with ganciclovir (27%; 95% CI, 25-29%; P < 0.001). In male patients who received HSCT from female donors, foscarnet recipients showed significantly lower incidence of extensive chronic GVHD than ganciclovir recipients, regardless of donor source or previous acute GVHD. While caution is necessary, these results indicate that foscarnet affects alloimmunization and might reduce the incidence of chronic GVHD.
{"title":"Association between early anti-cytomegalovirus therapy and the incidence of chronic graft-versus-host disease.","authors":"Kotaro Miyao, Makoto Murata, Tetsuya Nishida, Yukiyasu Ozawa, Naoyuki Uchida, Takahiro Fukuda, Noriko Doki, Tetsuya Eto, Toshiro Kawakita, Yasuo Mori, Satoru Takada, Hiroyuki Ohigashi, Masatsugu Tanaka, Yoshinobu Kanda, Ken-Ichi Matsuoka, Fumihiko Ishimaru, Yoshiko Atsuta, Junya Kanda, Seitaro Terakura","doi":"10.1007/s12185-024-03871-4","DOIUrl":"10.1007/s12185-024-03871-4","url":null,"abstract":"<p><p>Ganciclovir and foscarnet are two representative anti-cytomegalovirus (CMV) agents. A previous regional study revealed a lower risk of chronic graft-versus-host disease (GVHD) in patients who received pre-emptive foscarnet. We conducted a retrospective nationwide study to confirm the results. A total of 8890 patients aged 16 or older with hematological malignancies who received foscarnet (n = 1555) or ganciclovir (n = 7335) during their first hematopoietic stem cell transplantation (HSCT) were included. The risks of chronic GVHD (hazard ratio [HR], 1.26; 95% confidence interval [CI], 1.13-1.40; P < 0.001) and extensive chronic GVHD (HR, 1.16; 95% CI, 1.01-1.33; P = 0.033) were higher with ganciclovir. Among male patients with a female donor, the incidence of extensive chronic GVHD 3 years after HSCT was clearly lower with foscarnet (13%; 95% CI, 9-16%) than with ganciclovir (27%; 95% CI, 25-29%; P < 0.001). In male patients who received HSCT from female donors, foscarnet recipients showed significantly lower incidence of extensive chronic GVHD than ganciclovir recipients, regardless of donor source or previous acute GVHD. While caution is necessary, these results indicate that foscarnet affects alloimmunization and might reduce the incidence of chronic GVHD.</p>","PeriodicalId":13992,"journal":{"name":"International Journal of Hematology","volume":" ","pages":""},"PeriodicalIF":1.7,"publicationDate":"2024-11-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142619946","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Despite the introduction of new drugs, multiple myeloma (MM) still remains incurable. We previously reported that CD34+ MM cells, which are clonogenic and self-renewing, are therapy-resistant and persist as a major component of minimal residual disease, expanding during relapse. To investigate the effects of immunotherapies such as immune-checkpoint inhibitors, CAR-T therapy, and bispecific antibodies on CD34+ MM cells, we analyzed immune profiles of both MM cells and T cells from MM patients using microarrays and flow cytometry. Ingenuity pathway analysis revealed 14 out of 289 canonical pathways were more active in CD34+ MM cells compared to CD34- cells, many of which were involved in inflammation and immune responses. Notably, PD-1 signaling-related genes were highly expressed in CD34+ MM cells. Among 10 immune-checkpoint molecules, CD34+ cells more frequently expressed CD112, CD137L, CD270, CD275, and GAL9 than CD34- cells in both newly diagnosed and relapsed/resistant patients. In addition, CD4+ and CD8+ T cells more frequently expressed TIGIT and CD137, suggesting that CD112/TIGIT and CD137L/CD137 interactions may suppress T-cell activity against CD34+ MM cells. Furthermore, our finding of higher FcRH5 expression on CD34+ MM cells is encouraging for future research into the efficacy of FcRH5-targeted therapy in MM.
尽管新药层出不穷,但多发性骨髓瘤(MM)仍然无法治愈。我们以前曾报道过,CD34+ MM 细胞具有克隆性和自我更新性,对治疗具有抗药性,并且作为极小残留病的主要组成部分持续存在,并在复发时不断扩大。为了研究免疫检查点抑制剂、CAR-T疗法和双特异性抗体等免疫疗法对CD34+ MM细胞的影响,我们使用微阵列和流式细胞术分析了MM患者的MM细胞和T细胞的免疫特征。Ingenuity通路分析显示,与CD34-细胞相比,289条典型通路中有14条在CD34+ MM细胞中更为活跃,其中许多通路参与了炎症和免疫反应。值得注意的是,PD-1信号相关基因在CD34+ MM细胞中高度表达。在10种免疫检查点分子中,CD34+细胞比CD34-细胞更常表达CD112、CD137L、CD270、CD275和GAL9,无论是在新诊断患者还是复发/耐药患者中。此外,CD4+和CD8+ T细胞更频繁地表达TIGIT和CD137,这表明CD112/TIGIT和CD137L/CD137的相互作用可能会抑制T细胞对CD34+ MM细胞的活性。此外,我们在 CD34+ MM 细胞上发现了更高的 FcRH5 表达,这对未来研究 FcRH5 靶向治疗 MM 的疗效具有鼓舞作用。
{"title":"CD34<sup>+</sup> and CD34<sup>-</sup> MM cells show different immune-checkpoint molecule expression profiles: high expression of CD112 and CD137 ligand on CD34<sup>+</sup> MM cells.","authors":"Ayano Fukui-Morimoto, Kentaro Serizawa, Ko Fujimoto, Aki Hanamoto, Yoshio Iwata, Hiroaki Kakutani, Takahiro Kumode, Chikara Hirase, Yasuyoshi Morita, Yoichi Tatsumi, Hitoshi Hanamoto, Hirokazu Tanaka, Itaru Matsumura","doi":"10.1007/s12185-024-03867-0","DOIUrl":"https://doi.org/10.1007/s12185-024-03867-0","url":null,"abstract":"<p><p>Despite the introduction of new drugs, multiple myeloma (MM) still remains incurable. We previously reported that CD34<sup>+</sup> MM cells, which are clonogenic and self-renewing, are therapy-resistant and persist as a major component of minimal residual disease, expanding during relapse. To investigate the effects of immunotherapies such as immune-checkpoint inhibitors, CAR-T therapy, and bispecific antibodies on CD34<sup>+</sup> MM cells, we analyzed immune profiles of both MM cells and T cells from MM patients using microarrays and flow cytometry. Ingenuity pathway analysis revealed 14 out of 289 canonical pathways were more active in CD34<sup>+</sup> MM cells compared to CD34<sup>-</sup> cells, many of which were involved in inflammation and immune responses. Notably, PD-1 signaling-related genes were highly expressed in CD34<sup>+</sup> MM cells. Among 10 immune-checkpoint molecules, CD34<sup>+</sup> cells more frequently expressed CD112, CD137L, CD270, CD275, and GAL9 than CD34<sup>-</sup> cells in both newly diagnosed and relapsed/resistant patients. In addition, CD4<sup>+</sup> and CD8<sup>+</sup> T cells more frequently expressed TIGIT and CD137, suggesting that CD112/TIGIT and CD137L/CD137 interactions may suppress T-cell activity against CD34<sup>+</sup> MM cells. Furthermore, our finding of higher FcRH5 expression on CD34<sup>+</sup> MM cells is encouraging for future research into the efficacy of FcRH5-targeted therapy in MM.</p>","PeriodicalId":13992,"journal":{"name":"International Journal of Hematology","volume":" ","pages":""},"PeriodicalIF":1.7,"publicationDate":"2024-11-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142619948","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-11-11DOI: 10.1007/s12185-024-03868-z
Yoko Edahiro, Norio Komatsu
Polycythemia vera (PV) is a myeloproliferative neoplasm that is associated with an elevated risk of thrombosis. Treatment strategies are based on thrombosis risk classification. Phlebotomy is a commonly recommended treatment for all patients with PV, regardless of their risk classification, and reduces the incidence of thrombosis by lowering hematocrit levels. However, patients with PV frequently present with iron deficiency at diagnosis due to increased erythropoiesis, which repeated phlebotomy can exacerbate. This can produce symptoms that diminish quality of life, such as fatigue, lethargy, and impaired concentration. Recently, hepcidin mimetics have been developed to suppress iron utilization in erythropoiesis. Among them, rusfertide has been shown to control hematocrit levels without requiring phlebotomy. Further studies are needed to identify new treatment strategies for PV that also consider iron deficiency.
{"title":"Iron deficiency and phlebotomy in patients with polycythemia vera.","authors":"Yoko Edahiro, Norio Komatsu","doi":"10.1007/s12185-024-03868-z","DOIUrl":"https://doi.org/10.1007/s12185-024-03868-z","url":null,"abstract":"<p><p>Polycythemia vera (PV) is a myeloproliferative neoplasm that is associated with an elevated risk of thrombosis. Treatment strategies are based on thrombosis risk classification. Phlebotomy is a commonly recommended treatment for all patients with PV, regardless of their risk classification, and reduces the incidence of thrombosis by lowering hematocrit levels. However, patients with PV frequently present with iron deficiency at diagnosis due to increased erythropoiesis, which repeated phlebotomy can exacerbate. This can produce symptoms that diminish quality of life, such as fatigue, lethargy, and impaired concentration. Recently, hepcidin mimetics have been developed to suppress iron utilization in erythropoiesis. Among them, rusfertide has been shown to control hematocrit levels without requiring phlebotomy. Further studies are needed to identify new treatment strategies for PV that also consider iron deficiency.</p>","PeriodicalId":13992,"journal":{"name":"International Journal of Hematology","volume":" ","pages":""},"PeriodicalIF":1.7,"publicationDate":"2024-11-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142619950","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Successful complete remission after induction therapy with CPX-351 for acute myeloid leukemia with myelodysplasia-related changes accompanied by double-minute chromosomes and MYC gene amplification.","authors":"Masuho Saburi, Takumi Nishikawa, Katsuya Kawano, Eiichi Ohtsuka","doi":"10.1007/s12185-024-03876-z","DOIUrl":"https://doi.org/10.1007/s12185-024-03876-z","url":null,"abstract":"","PeriodicalId":13992,"journal":{"name":"International Journal of Hematology","volume":" ","pages":""},"PeriodicalIF":1.7,"publicationDate":"2024-11-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142619954","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
The advent of tyrosine kinase inhibitors (TKIs) has revolutionized the treatment of chronic myeloid leukemia (CML), significantly improving patient prognosis. Asciminib, a novel specifically targeting the ABL myristoyl pocket inhibitor, has shown promise for CML patients unresponsive or intolerant to traditional TKIs. However, its use in hemodialysis patients remains underexplored. We present a case of a 71-year-old man with CML undergoing hemodialysis, successfully treated with asciminib. Initial treatment with bosutinib was effective but later failed, prompting a switch to asciminib. The patient achieved a major molecular response within 2 months without adverse effects. Pharmacokinetic analysis revealed significant drug clearance during hemodialysis, necessitating dosage adjustments. This case highlights the potential of asciminib in managing CML in hemodialysis patients, emphasizing the need for individualized treatment plans and close monitoring. Further studies are warranted to establish comprehensive guidelines for asciminib use in this unique patient population.
{"title":"Safety and efficacy of asciminib in a patient with chronic myeloid leukemia on hemodialysis.","authors":"Ryosuke Naka, Yoshimitsu Shimomura, Masatomo Miura, Yuya Nagai, Tadakazu Kondo, Takayuki Ishikawa","doi":"10.1007/s12185-024-03869-y","DOIUrl":"https://doi.org/10.1007/s12185-024-03869-y","url":null,"abstract":"<p><p>The advent of tyrosine kinase inhibitors (TKIs) has revolutionized the treatment of chronic myeloid leukemia (CML), significantly improving patient prognosis. Asciminib, a novel specifically targeting the ABL myristoyl pocket inhibitor, has shown promise for CML patients unresponsive or intolerant to traditional TKIs. However, its use in hemodialysis patients remains underexplored. We present a case of a 71-year-old man with CML undergoing hemodialysis, successfully treated with asciminib. Initial treatment with bosutinib was effective but later failed, prompting a switch to asciminib. The patient achieved a major molecular response within 2 months without adverse effects. Pharmacokinetic analysis revealed significant drug clearance during hemodialysis, necessitating dosage adjustments. This case highlights the potential of asciminib in managing CML in hemodialysis patients, emphasizing the need for individualized treatment plans and close monitoring. Further studies are warranted to establish comprehensive guidelines for asciminib use in this unique patient population.</p>","PeriodicalId":13992,"journal":{"name":"International Journal of Hematology","volume":" ","pages":""},"PeriodicalIF":1.7,"publicationDate":"2024-11-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142604098","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-11-06DOI: 10.1007/s12185-024-03840-x
Masashi Sawa, Toshihiro Miyamoto, Hee-Je Kim, Yasushi Hiramatsu, June-Won Cheong, Takayuki Ikezoe, Tomoki Naoe, Koichi Akashi, Satoshi Morita, Masanori Kosako, Moyu Ikegaya, Wataru Terada, Takeshi Kadokura, Jason Hill, Shuichi Miyawaki, Stanley C Gill, Alexandra Heinloth, Nahla Hasabou
Objective: This interim analysis of a phase 1/2, open-label, single-arm study assessed the safety, efficacy, and pharmacokinetics of gilteritinib plus chemotherapy in adults with newly diagnosed FLT3 mutation-positive acute myeloid leukemia.
Methods: In sequential phase 1 and 2 studies, induction and consolidation therapy with gilteritinib 120 mg/day plus chemotherapy (induction: idarubicin/cytarabine once daily; consolidation: cytarabine twice daily) was followed by maintenance gilteritinib 120 mg/day monotherapy. Endpoints included maximum tolerated dose (MTD), recommended expansion dose (RED), and dose-limiting toxicity (phase 1), and complete remission (CR) rate following induction therapy (primary endpoint), overall survival (OS), safety, and pharmacokinetics (phase 2).
Results: In phase 1, MTD was not reached and RED was 120 mg/day. In phase 2, the CR rate was 50.0% after induction (90% confidence interval [CI] 40.4, 59.6); however, the lower confidence limit did not exceed the pre-defined 55% benchmark. Composite CR (CRc) rates were high following induction (86.6%, 95% CI [77.3, 93.1]), consolidation, and maintenance therapy (87.8%, 95% CI [78.7, 94.0], each). The probability of OS was 86.6% at 12 months. No new safety findings were reported.
Conclusion: In this interim analysis, gilteritinib 120 mg/day in combination with chemotherapy was well tolerated, with similar CRc rates to previous studies.
{"title":"A phase 1/2 study of gilteritinib in combination with chemotherapy in newly diagnosed patients with AML in Asia.","authors":"Masashi Sawa, Toshihiro Miyamoto, Hee-Je Kim, Yasushi Hiramatsu, June-Won Cheong, Takayuki Ikezoe, Tomoki Naoe, Koichi Akashi, Satoshi Morita, Masanori Kosako, Moyu Ikegaya, Wataru Terada, Takeshi Kadokura, Jason Hill, Shuichi Miyawaki, Stanley C Gill, Alexandra Heinloth, Nahla Hasabou","doi":"10.1007/s12185-024-03840-x","DOIUrl":"https://doi.org/10.1007/s12185-024-03840-x","url":null,"abstract":"<p><strong>Objective: </strong>This interim analysis of a phase 1/2, open-label, single-arm study assessed the safety, efficacy, and pharmacokinetics of gilteritinib plus chemotherapy in adults with newly diagnosed FLT3 mutation-positive acute myeloid leukemia.</p><p><strong>Methods: </strong>In sequential phase 1 and 2 studies, induction and consolidation therapy with gilteritinib 120 mg/day plus chemotherapy (induction: idarubicin/cytarabine once daily; consolidation: cytarabine twice daily) was followed by maintenance gilteritinib 120 mg/day monotherapy. Endpoints included maximum tolerated dose (MTD), recommended expansion dose (RED), and dose-limiting toxicity (phase 1), and complete remission (CR) rate following induction therapy (primary endpoint), overall survival (OS), safety, and pharmacokinetics (phase 2).</p><p><strong>Results: </strong>In phase 1, MTD was not reached and RED was 120 mg/day. In phase 2, the CR rate was 50.0% after induction (90% confidence interval [CI] 40.4, 59.6); however, the lower confidence limit did not exceed the pre-defined 55% benchmark. Composite CR (CRc) rates were high following induction (86.6%, 95% CI [77.3, 93.1]), consolidation, and maintenance therapy (87.8%, 95% CI [78.7, 94.0], each). The probability of OS was 86.6% at 12 months. No new safety findings were reported.</p><p><strong>Conclusion: </strong>In this interim analysis, gilteritinib 120 mg/day in combination with chemotherapy was well tolerated, with similar CRc rates to previous studies.</p>","PeriodicalId":13992,"journal":{"name":"International Journal of Hematology","volume":" ","pages":""},"PeriodicalIF":1.7,"publicationDate":"2024-11-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142582860","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
The Japan Children's Cancer Group Relapsed Acute Lymphoblastic Leukemia (ALL) Committee conducted a prospective observational study (ALL-R14) to explore promising reinduction therapy regimens for relapsed ALL to investigate in future trials. In Japan, clofarabine- and bortezomib-based regimens were of interest since they were newly introduced for ALL in the study period (2015-2018). Seventy-five pediatric patients were enrolled in total. The 2-year event-free/overall survival rates in patients with first (n = 59) or second (n = 11) relapse were 40.1% (95% confidence interval [CI]: 25.5-52.3%)/66.3% (95% CI 52.3-77.0%) and 34.1% (95% CI 9.1-61.6%)/62.3% (95% CI 27.7-84.0%), respectively. Clofarabine- or bortezomib-based regimens were used only in patients with high-risk disease. The first reinduction therapy used in the 41 patients with early or multiple relapsed B-cell precursor ALL was clofarabine in 7 patients and bortezomib in 9 patients. The odds ratio for reinduction failure risk with a clofarabine- or bortezomib-based regimen compared with other regimens was 9.0 (95% CI 0.9-86.4, P = 0.057) or 1.9 (95% CI 0.4-8.7, P = 0.42), respectively. Thus, clofarabine- or bortezomib-based regimens had no obvious advantage as reinduction therapy for relapsed ALL in children.
日本儿童癌症小组复发性急性淋巴细胞白血病(ALL)委员会开展了一项前瞻性观察研究(ALL-R14),旨在探索有前景的复发性ALL再诱导治疗方案,以便在未来的试验中进行研究。在日本,基于氯法拉滨和硼替佐米的治疗方案是研究期间(2015-2018 年)新引入的 ALL 治疗方案,因此备受关注。共有75名儿科患者入组。首次(n = 59)或第二次(n = 11)复发患者的2年无事件/总生存率分别为40.1%(95%置信区间[CI]:25.5-52.3%)/66.3%(95% CI 52.3-77.0%)和34.1%(95% CI 9.1-61.6%)/62.3%(95% CI 27.7-84.0%)。氯法拉滨或硼替佐米方案仅用于高危患者。在41例早期或多次复发的B细胞前体ALL患者中,7例患者的首次恢复疗法为氯法拉滨,9例患者的首次恢复疗法为硼替佐米。与其他方案相比,氯法拉滨或硼替佐米方案的再诱导失败风险几率比分别为9.0(95% CI 0.9-86.4,P = 0.057)或1.9(95% CI 0.4-8.7,P = 0.42)。因此,以氯法拉滨或硼替佐米为基础的方案作为复发儿童 ALL 的还原疗法没有明显优势。
{"title":"Treatment of relapsed acute lymphoblastic leukemia in children: an observational study of the Japan Children's Cancer Group.","authors":"Hiroaki Goto, Akiko Kada, Chitose Ogawa, Ritsuo Nishiuchi, Junko Yamanaka, Akihiro Iguchi, Masanori Nishi, Kimiyoshi Sakaguchi, Tadashi Kumamoto, Shinji Mochizuki, Hideaki Ueki, Yoshiyuki Kosaka, Akiko M Saito, Hidemi Toyoda","doi":"10.1007/s12185-024-03838-5","DOIUrl":"10.1007/s12185-024-03838-5","url":null,"abstract":"<p><p>The Japan Children's Cancer Group Relapsed Acute Lymphoblastic Leukemia (ALL) Committee conducted a prospective observational study (ALL-R14) to explore promising reinduction therapy regimens for relapsed ALL to investigate in future trials. In Japan, clofarabine- and bortezomib-based regimens were of interest since they were newly introduced for ALL in the study period (2015-2018). Seventy-five pediatric patients were enrolled in total. The 2-year event-free/overall survival rates in patients with first (n = 59) or second (n = 11) relapse were 40.1% (95% confidence interval [CI]: 25.5-52.3%)/66.3% (95% CI 52.3-77.0%) and 34.1% (95% CI 9.1-61.6%)/62.3% (95% CI 27.7-84.0%), respectively. Clofarabine- or bortezomib-based regimens were used only in patients with high-risk disease. The first reinduction therapy used in the 41 patients with early or multiple relapsed B-cell precursor ALL was clofarabine in 7 patients and bortezomib in 9 patients. The odds ratio for reinduction failure risk with a clofarabine- or bortezomib-based regimen compared with other regimens was 9.0 (95% CI 0.9-86.4, P = 0.057) or 1.9 (95% CI 0.4-8.7, P = 0.42), respectively. Thus, clofarabine- or bortezomib-based regimens had no obvious advantage as reinduction therapy for relapsed ALL in children.</p>","PeriodicalId":13992,"journal":{"name":"International Journal of Hematology","volume":" ","pages":"631-638"},"PeriodicalIF":1.7,"publicationDate":"2024-11-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142072783","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}