Semaglutide impairs bioavailability of alectinib: a note of warning based on a cross-over pharmacokinetic drug-drug interaction study

IF 24.9 1区 医学 Q1 ONCOLOGY
Niels Heersche, Daan A. C. Lanser, Esther Oomen-de Hoop, Attila Içli, Peter de Bruijn, Marthe S. Paats, Elisabeth F. C. van Rossum, Stijn L. W. Koolen, Ron H. N. van Schaik, Anne-Marie C. Dingemans, G. D. Marijn Veerman, Ron H. J. Mathijssen
{"title":"Semaglutide impairs bioavailability of alectinib: a note of warning based on a cross-over pharmacokinetic drug-drug interaction study","authors":"Niels Heersche,&nbsp;Daan A. C. Lanser,&nbsp;Esther Oomen-de Hoop,&nbsp;Attila Içli,&nbsp;Peter de Bruijn,&nbsp;Marthe S. Paats,&nbsp;Elisabeth F. C. van Rossum,&nbsp;Stijn L. W. Koolen,&nbsp;Ron H. N. van Schaik,&nbsp;Anne-Marie C. Dingemans,&nbsp;G. D. Marijn Veerman,&nbsp;Ron H. J. Mathijssen","doi":"10.1002/cac2.70030","DOIUrl":null,"url":null,"abstract":"<p>Alectinib is a first-line treatment for patients with advanced non-small cell lung cancer (NSCLC) harboring anaplastic lymphoma kinase-positive (<i>ALK+</i>) driver aberrations with a median progression-free survival of 35 months and a 5-year overall survival of 63% [<span>1</span>]. Currently, alectinib also shows improvement as an adjuvant treatment in resected stage IA-IIIB <i>ALK+</i> NSCLC [<span>2</span>]. Alectinib has a mild safety profile, but a notable underreported side-effect is weight gain [<span>3</span>]. Studies show sarcopenic obesity rates doubling from 24% to 47% in the first year of treatment [<span>3</span>], with persisting weight gain appearing early [<span>4</span>]. Given patients’ extended survival, this poses risks for metabolic, cardiovascular, and psychological health.</p><p>Interestingly, over the past few years, glucagon-like peptide 1 (GLP-1) receptor agonists semaglutide, liraglutide, and tirzepatide, have been approved as promising anti-obesity drugs [<span>5</span>]. Subcutaneous, once-weekly semaglutide induces a weight loss of 15% after 68 weeks [<span>5</span>]. Hence, semaglutide might pose an interesting means of treating alectinib-induced weight gain. Recently, it was reported that a patient who experienced 20 kg weight gain during treatment with alectinib and lorlatinib, achieved 5 kg weight loss in just 6 months with semaglutide [<span>6</span>].</p><p>In a retrospective analysis, alectinib plasma trough levels above 435 ng/mL correlated with prolonged effectiveness compared to lower exposures [<span>7</span>]. Despite moderate interpatient variability of 40%-45%, the lipophilicity of alectinib makes it highly dependent on dietary fat for sufficient dissolution and subsequent absorption in the gastro-intestinal tract to maintain adequate trough concentrations [<span>7, 8</span>]. Considering that semaglutide decreases appetite, patients may inadvertently decrease their dietary (fat) intake, hampering absorption of alectinib. Hence, we investigated the effects of semaglutide on the pharmacokinetics of alectinib to gain insight into the pharmacokinetic interplay between both.</p><p>Therefore, we included 10 patients in a two-period cross-over study comparing alectinib exposure on alectinib monotherapy (period A) versus co-administration of semaglutide (period B) (Supplementary Table S1). Each treatment period lasted one week. Semaglutide was administrated as a single dose of 2.0 mg subcutaneously. Further details on the methodology of this study and baseline patient characteristics are provided in the Supplementary Materials.</p><p>After alectinib monotherapy (period A), a geometric mean of the area under the curve (AUC<sub>0-10h</sub>) of 7,114 ng × h/mL (coefficient of variation [CV] = 34%) was observed, compared to an AUC<sub>0-10h</sub> of 4,843 ng × h/mL (CV = 47%) after co-administering alectinib with subcutaneous semaglutide (period B). This change in alectinib geometric mean AUC<sub>0-10h</sub> constituted a significant and clinically relevant reduction of 32% (95% confidence interval [CI] = -45% to -15%; <i>P</i> = 0.004; Figure 1A and Supplementary Table S2) when co-administering semaglutide. Additionally, trough concentrations (C<sub>trough</sub>) showed a similar trend towards lower concentrations with semaglutide, decreasing the geometric mean C<sub>trough</sub> from 681 ng/mL (CV = 29%) to 509 ng/mL (CV = 66%), reflecting a relative difference of -25% (95% CI = -46% to 3%; <i>P</i> = 0.072) when alectinib was combined with semaglutide (period B) compared to alectinib alone (period A). The maximum concentration (C<sub>max</sub>) decreased with 36% (95% CI = -48% to -20%; <i>P</i> = 0.001) with a C<sub>max</sub> of alectinib of 875 ng/mL (CV = 31%) on monotherapy (period A) versus 563 ng/mL (CV = 42%) in the combination arm (period B).</p><p>Moreover, during alectinib monotherapy, all patients had C<sub>trough</sub> levels surpassing the efficacy threshold (i.e. &gt;435 ng/mL) [<span>7</span>], whilst subsequent to the combination of alectinib and semaglutide, only 60% retained plasma levels above the efficacy threshold (<i>P</i> = 0.125), see Supplementary Table S3. Plasma samples taken after trial completion demonstrated that in all but 2 patients, C<sub>trough</sub> levels returned to above the efficacy threshold (i.e. &gt;435 ng/mL) at follow-up visit 1, and only 1 patient remained below the threshold at follow-up visit 2 (Figure 1B).</p><p>After the co-administration of semaglutide, patients experienced substantially more toxicity compared to alectinib monotherapy. Gastro-intestinal side-effects such as vomiting (period A versus period B: 0% versus 80%), nausea (period A versus period B: 0% versus 50%), and anorexia (period A versus period B: 0% versus 50%) were more prevalent after administration of semaglutide. All toxicities encompassed grade 1 or 2 toxicity according to the Common Terminology Criteria for Adverse Events (CTCAE v5.0), see Supplementary Table S4. Review of the patient's diaries revealed a tendency to lower food intake after semaglutide administration.</p><p>As this is the first study to describe an interaction between an anti-cancer agent and semaglutide, these findings hold importance for clinicians and cancer patients. As mentioned, for alectinib an exposure-response relationship has been established based on a retrospective analysis [<span>7</span>]. The value of this threshold for progression free survival, is currently studied in a prospective randomized trial; the so-called ADAPT-ALEC study which is accruing patients in the Netherlands and France (NCT05525338) [<span>7</span>]. Given this assumed exposure-response relationship, combining alectinib and semaglutide could potentially hamper the anti-neoplastic efficacy of alectinib. In our study, 4 out of 10 subjects had trough levels below the 435 ng/mL threshold after the combination of semaglutide and alectinib, compared to none on alectinib monotherapy. All four of these patients were dosed alectinib 450 mg twice daily (BID).</p><p>The mechanism of action behind GLP-1 receptor agonists is manifold. In part, the positive effect on weight loss might be due to the induction of satiety by mimicking GLP-1, thereby causing loss of appetite [<span>5</span>]. In addition, GLP-1 receptor agonists reduce gastric motility, resulting in a prolonged sensation of repletion after ingestion of food [<span>5</span>]. This mechanism also causes gastro-intestinal side-effects, such as vomiting. In our trial, nausea and vomiting led to at least one missed dose of alectinib in 5 patients, potentially contributing to the reduction in alectinib AUC<sub>0-10h</sub>. Nonetheless, most side-effects were transient and only occurred within the first days post-semaglutide administration (Supplementary Figure S1). Notably, all but one missed alectinib dose occurred during the first two days of period B, indicating all patients should have reached steady state concentrations by the time of pharmacokinetic sampling. Additionally, post-hoc sensitivity analysis found no effect of the missed doses on alectinib exposure (Supplementary Table S5). Conversely, a food effect might provide a more plausible hypothesis for the observed reduced alectinib exposure after semaglutide administration. Notably, most patients in our study struggled to adhere to the prescribed diet due to loss of appetite and gastrointestinal adverse events, which are well-known side-effects of semaglutide. Since alectinib is administered orally, its absorption is significantly influenced by food intake [<span>8</span>], in particular since the bioavailability is low (37%), even under fed conditions. Therefore, alterations in food intake will have significant impact on the systemic alectinib exposure. In our study, food (and hence fat) intake was impaired in most patients due to loss of appetite, likely compromising alectinib absorption [<span>8</span>]. This is especially relevant since another study found that patients on semaglutide report having less preference for foods high in fat [<span>9</span>].</p><p>In our trial, semaglutide was administered once to assess pharmacokinetic safety. Based on our results, we highlight the need for caution when prescribing semaglutide, especially in alectinib dose-reduced patients. While semaglutide still remains a promising therapy for alectinib-induced weight gain, further research is necessary to evaluate long-term effects of semaglutide on alectinib treatment. For instance, weight loss itself is associated with lower alectinib clearance, which could subsequently increase exposure in the long-term [<span>10</span>].</p><p>In conclusion, our study highlights a clinically relevant pharmacokinetic drug-drug interaction between semaglutide and alectinib resulting in a major decrease in alectinib exposure, most likely by a semaglutide-mediated food effect. This underscores the importance of exercising caution when prescribing alectinib and semaglutide (and potentially other GLP-1 receptor agonists) concurrently, and necessitates monitoring of alectinib plasma concentrations in these patients.</p><p><i>Study concept and design</i>: Niels Heersche, Daan A.C. Lanser, Stijn L.W. Koolen, Anne-Marie C. Dingemans, G.D. Marijn Veerman, and Ron H.J. Mathijssen. <i>Study performance</i>: Niels Heersche, Daan A.C. Lanser, Attila Icli, and Peter de Bruijn. <i>Patient selection</i>: Marthe S. Paats and Anne-Marie C. Dingemans. <i>Data analysis and interpretation</i>: Niels Heersche, Daan A.C. Lanser, Esther Oomen-de Hoop, Attila Icli, G.D. Marijn Veerman, and Ron H.J. Mathijssen. <i>Writing—original draft</i>: Niels Heersche, Daan A.C. Lanser, G.D. Marijn Veerman, and Ron H.J. Mathijssen. <i>Writing—review &amp; editing</i>: Niels Heersche, Daan A.C. Lanser, Esther Oomen-de Hoop, Attila Icli, Peter de Bruijn, Marthe S. Paats, Elisabeth F.C. van Rossum, Stijn L.W. Koolen, Ron H.N. van Schaik, Anne-Marie C. Dingemans, G.D. Marijn Veerman, and Ron H.J. Mathijssen.</p><p>EFCvR is involved in clinical trials with Rhythm Pharmaceuticals for targeted therapy for rare genetic obesity (all paid to the institute). ACD reports grants (all paid to the institute) from Amgen. RHJM reports unrestricted grants for investigator-initiated trials (all paid to the institute) from Astellas, Bayer, Boehringer-Ingelheim, Cristal Therapeutics, Deuter Oncology, Echo Pharmaceuticals, Nordic Pharma, Novartis, Pamgene, Pfizer, Roche, Sanofi, and Servier. None of the other authors reports COIs.</p><p>Not applicable.</p><p>The competent authority and the local ethics committee of the Erasmus University Medical Center Rotterdam approved the trial (registration ID: NL78079.078.23 and MEC 21-0478, respectively). All participants provided written informed consent prior to their inclusion in the study.</p><p>Dutch Trial Registry ID: NL9702</p>","PeriodicalId":9495,"journal":{"name":"Cancer Communications","volume":"45 8","pages":"914-918"},"PeriodicalIF":24.9000,"publicationDate":"2025-05-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/cac2.70030","citationCount":"0","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"Cancer Communications","FirstCategoryId":"3","ListUrlMain":"https://onlinelibrary.wiley.com/doi/10.1002/cac2.70030","RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q1","JCRName":"ONCOLOGY","Score":null,"Total":0}
引用次数: 0

Abstract

Alectinib is a first-line treatment for patients with advanced non-small cell lung cancer (NSCLC) harboring anaplastic lymphoma kinase-positive (ALK+) driver aberrations with a median progression-free survival of 35 months and a 5-year overall survival of 63% [1]. Currently, alectinib also shows improvement as an adjuvant treatment in resected stage IA-IIIB ALK+ NSCLC [2]. Alectinib has a mild safety profile, but a notable underreported side-effect is weight gain [3]. Studies show sarcopenic obesity rates doubling from 24% to 47% in the first year of treatment [3], with persisting weight gain appearing early [4]. Given patients’ extended survival, this poses risks for metabolic, cardiovascular, and psychological health.

Interestingly, over the past few years, glucagon-like peptide 1 (GLP-1) receptor agonists semaglutide, liraglutide, and tirzepatide, have been approved as promising anti-obesity drugs [5]. Subcutaneous, once-weekly semaglutide induces a weight loss of 15% after 68 weeks [5]. Hence, semaglutide might pose an interesting means of treating alectinib-induced weight gain. Recently, it was reported that a patient who experienced 20 kg weight gain during treatment with alectinib and lorlatinib, achieved 5 kg weight loss in just 6 months with semaglutide [6].

In a retrospective analysis, alectinib plasma trough levels above 435 ng/mL correlated with prolonged effectiveness compared to lower exposures [7]. Despite moderate interpatient variability of 40%-45%, the lipophilicity of alectinib makes it highly dependent on dietary fat for sufficient dissolution and subsequent absorption in the gastro-intestinal tract to maintain adequate trough concentrations [7, 8]. Considering that semaglutide decreases appetite, patients may inadvertently decrease their dietary (fat) intake, hampering absorption of alectinib. Hence, we investigated the effects of semaglutide on the pharmacokinetics of alectinib to gain insight into the pharmacokinetic interplay between both.

Therefore, we included 10 patients in a two-period cross-over study comparing alectinib exposure on alectinib monotherapy (period A) versus co-administration of semaglutide (period B) (Supplementary Table S1). Each treatment period lasted one week. Semaglutide was administrated as a single dose of 2.0 mg subcutaneously. Further details on the methodology of this study and baseline patient characteristics are provided in the Supplementary Materials.

After alectinib monotherapy (period A), a geometric mean of the area under the curve (AUC0-10h) of 7,114 ng × h/mL (coefficient of variation [CV] = 34%) was observed, compared to an AUC0-10h of 4,843 ng × h/mL (CV = 47%) after co-administering alectinib with subcutaneous semaglutide (period B). This change in alectinib geometric mean AUC0-10h constituted a significant and clinically relevant reduction of 32% (95% confidence interval [CI] = -45% to -15%; P = 0.004; Figure 1A and Supplementary Table S2) when co-administering semaglutide. Additionally, trough concentrations (Ctrough) showed a similar trend towards lower concentrations with semaglutide, decreasing the geometric mean Ctrough from 681 ng/mL (CV = 29%) to 509 ng/mL (CV = 66%), reflecting a relative difference of -25% (95% CI = -46% to 3%; P = 0.072) when alectinib was combined with semaglutide (period B) compared to alectinib alone (period A). The maximum concentration (Cmax) decreased with 36% (95% CI = -48% to -20%; P = 0.001) with a Cmax of alectinib of 875 ng/mL (CV = 31%) on monotherapy (period A) versus 563 ng/mL (CV = 42%) in the combination arm (period B).

Moreover, during alectinib monotherapy, all patients had Ctrough levels surpassing the efficacy threshold (i.e. >435 ng/mL) [7], whilst subsequent to the combination of alectinib and semaglutide, only 60% retained plasma levels above the efficacy threshold (P = 0.125), see Supplementary Table S3. Plasma samples taken after trial completion demonstrated that in all but 2 patients, Ctrough levels returned to above the efficacy threshold (i.e. >435 ng/mL) at follow-up visit 1, and only 1 patient remained below the threshold at follow-up visit 2 (Figure 1B).

After the co-administration of semaglutide, patients experienced substantially more toxicity compared to alectinib monotherapy. Gastro-intestinal side-effects such as vomiting (period A versus period B: 0% versus 80%), nausea (period A versus period B: 0% versus 50%), and anorexia (period A versus period B: 0% versus 50%) were more prevalent after administration of semaglutide. All toxicities encompassed grade 1 or 2 toxicity according to the Common Terminology Criteria for Adverse Events (CTCAE v5.0), see Supplementary Table S4. Review of the patient's diaries revealed a tendency to lower food intake after semaglutide administration.

As this is the first study to describe an interaction between an anti-cancer agent and semaglutide, these findings hold importance for clinicians and cancer patients. As mentioned, for alectinib an exposure-response relationship has been established based on a retrospective analysis [7]. The value of this threshold for progression free survival, is currently studied in a prospective randomized trial; the so-called ADAPT-ALEC study which is accruing patients in the Netherlands and France (NCT05525338) [7]. Given this assumed exposure-response relationship, combining alectinib and semaglutide could potentially hamper the anti-neoplastic efficacy of alectinib. In our study, 4 out of 10 subjects had trough levels below the 435 ng/mL threshold after the combination of semaglutide and alectinib, compared to none on alectinib monotherapy. All four of these patients were dosed alectinib 450 mg twice daily (BID).

The mechanism of action behind GLP-1 receptor agonists is manifold. In part, the positive effect on weight loss might be due to the induction of satiety by mimicking GLP-1, thereby causing loss of appetite [5]. In addition, GLP-1 receptor agonists reduce gastric motility, resulting in a prolonged sensation of repletion after ingestion of food [5]. This mechanism also causes gastro-intestinal side-effects, such as vomiting. In our trial, nausea and vomiting led to at least one missed dose of alectinib in 5 patients, potentially contributing to the reduction in alectinib AUC0-10h. Nonetheless, most side-effects were transient and only occurred within the first days post-semaglutide administration (Supplementary Figure S1). Notably, all but one missed alectinib dose occurred during the first two days of period B, indicating all patients should have reached steady state concentrations by the time of pharmacokinetic sampling. Additionally, post-hoc sensitivity analysis found no effect of the missed doses on alectinib exposure (Supplementary Table S5). Conversely, a food effect might provide a more plausible hypothesis for the observed reduced alectinib exposure after semaglutide administration. Notably, most patients in our study struggled to adhere to the prescribed diet due to loss of appetite and gastrointestinal adverse events, which are well-known side-effects of semaglutide. Since alectinib is administered orally, its absorption is significantly influenced by food intake [8], in particular since the bioavailability is low (37%), even under fed conditions. Therefore, alterations in food intake will have significant impact on the systemic alectinib exposure. In our study, food (and hence fat) intake was impaired in most patients due to loss of appetite, likely compromising alectinib absorption [8]. This is especially relevant since another study found that patients on semaglutide report having less preference for foods high in fat [9].

In our trial, semaglutide was administered once to assess pharmacokinetic safety. Based on our results, we highlight the need for caution when prescribing semaglutide, especially in alectinib dose-reduced patients. While semaglutide still remains a promising therapy for alectinib-induced weight gain, further research is necessary to evaluate long-term effects of semaglutide on alectinib treatment. For instance, weight loss itself is associated with lower alectinib clearance, which could subsequently increase exposure in the long-term [10].

In conclusion, our study highlights a clinically relevant pharmacokinetic drug-drug interaction between semaglutide and alectinib resulting in a major decrease in alectinib exposure, most likely by a semaglutide-mediated food effect. This underscores the importance of exercising caution when prescribing alectinib and semaglutide (and potentially other GLP-1 receptor agonists) concurrently, and necessitates monitoring of alectinib plasma concentrations in these patients.

Study concept and design: Niels Heersche, Daan A.C. Lanser, Stijn L.W. Koolen, Anne-Marie C. Dingemans, G.D. Marijn Veerman, and Ron H.J. Mathijssen. Study performance: Niels Heersche, Daan A.C. Lanser, Attila Icli, and Peter de Bruijn. Patient selection: Marthe S. Paats and Anne-Marie C. Dingemans. Data analysis and interpretation: Niels Heersche, Daan A.C. Lanser, Esther Oomen-de Hoop, Attila Icli, G.D. Marijn Veerman, and Ron H.J. Mathijssen. Writing—original draft: Niels Heersche, Daan A.C. Lanser, G.D. Marijn Veerman, and Ron H.J. Mathijssen. Writing—review & editing: Niels Heersche, Daan A.C. Lanser, Esther Oomen-de Hoop, Attila Icli, Peter de Bruijn, Marthe S. Paats, Elisabeth F.C. van Rossum, Stijn L.W. Koolen, Ron H.N. van Schaik, Anne-Marie C. Dingemans, G.D. Marijn Veerman, and Ron H.J. Mathijssen.

EFCvR is involved in clinical trials with Rhythm Pharmaceuticals for targeted therapy for rare genetic obesity (all paid to the institute). ACD reports grants (all paid to the institute) from Amgen. RHJM reports unrestricted grants for investigator-initiated trials (all paid to the institute) from Astellas, Bayer, Boehringer-Ingelheim, Cristal Therapeutics, Deuter Oncology, Echo Pharmaceuticals, Nordic Pharma, Novartis, Pamgene, Pfizer, Roche, Sanofi, and Servier. None of the other authors reports COIs.

Not applicable.

The competent authority and the local ethics committee of the Erasmus University Medical Center Rotterdam approved the trial (registration ID: NL78079.078.23 and MEC 21-0478, respectively). All participants provided written informed consent prior to their inclusion in the study.

Dutch Trial Registry ID: NL9702

Abstract Image

西马鲁肽损害阿勒替尼的生物利用度:一项基于交叉药代动力学药物相互作用研究的警告。
Alectinib是晚期非小细胞肺癌(NSCLC)患者的一线治疗药物,该患者携带间变性淋巴瘤激酶阳性(ALK+)驱动异常,中位无进展生存期为35个月,5年总生存期为63%。目前,alectinib作为切除的IA-IIIB期ALK+ NSCLC[2]的辅助治疗也显示出改善。Alectinib具有轻微的安全性,但一个值得注意的未被报道的副作用是体重增加。研究表明,在治疗的第一年,肌肉减少型肥胖率翻了一番,从24%增加到47%,体重持续增加,出现在[3]早期。鉴于患者的生存期延长,这对代谢、心血管和心理健康构成了风险。有趣的是,在过去几年中,胰高血糖素样肽1 (GLP-1)受体激动剂semaglutide, liraglutide和tizepatide已被批准为有前景的抗肥胖药物[5]。皮下注射每周一次的西马鲁肽,68周后体重减轻15%。因此,西马鲁肽可能是治疗阿勒替尼引起的体重增加的一种有趣的方法。最近有报道称,一名患者在使用阿勒替尼和氯拉替尼治疗期间体重增加了20公斤,但在使用西马鲁肽[6]的短短6个月内体重减轻了5公斤。在回顾性分析中,与较低暴露水平相比,高于435 ng/mL的阿勒替尼血浆谷水平与长效相关。尽管患者间的差异为40%-45%,但alectinib的亲脂性使其高度依赖膳食脂肪来充分溶解和随后在胃肠道中的吸收,以维持足够的谷浓度[7,8]。考虑到西马鲁肽会降低食欲,患者可能会无意中减少他们的饮食(脂肪)摄入量,阻碍了阿勒替尼的吸收。因此,我们研究了西马鲁肽对阿勒替尼药代动力学的影响,以深入了解两者之间的药代动力学相互作用。因此,我们纳入了10例患者进行两期交叉研究,比较阿勒替尼单药治疗(a期)与西马鲁肽联合治疗(B期)(补充表S1)。每组治疗1周。西马鲁肽单次皮下给药2.0 mg。关于本研究方法学和基线患者特征的进一步细节请参见补充资料。在阿勒替尼单药治疗(A期)后,观察到曲线下面积(AUC0-10h)的几何平均值为7,114 ng × h/mL(变异系数[CV] = 34%),而在阿勒替尼与皮下塞马鲁肽联合使用(B期)后,AUC0-10h为4,843 ng × h/mL (CV = 47%)。当联合使用西马鲁肽时,阿勒替尼几何平均AUC0-10h的变化构成了32%的显著和临床相关的减少(95%置信区间[CI] = -45%至-15%;P = 0.004;图1A和补充表S2)。此外,谷浓度(Ctrough)也显示出类似的降低浓度的趋势,几何平均谷浓度从681 ng/mL (CV = 29%)降低到509 ng/mL (CV = 66%),反映了与阿勒替尼单独(a期)相比,阿勒替尼联合(B期)与阿勒替尼(a期)的相对差异为-25% (95% CI = -46%至3%;P = 0.072)。最大浓度(Cmax)下降了36% (95% CI = -48%至-20%;P = 0.001),单药组(a期)的Cmax为875 ng/mL (CV = 31%),而联合治疗组(B期)的Cmax为563 ng/mL (CV = 42%)。此外,在阿勒替尼单药治疗期间,所有患者的血浆水平均超过了有效阈值(即&gt;435 ng/mL)[7],而在阿勒替尼和西马鲁肽联合治疗后,只有60%的患者血浆水平保持在有效阈值以上(P = 0.125),见补充表S3。试验结束后采集的血浆样本显示,除2例患者外,其余患者在随访1时,cough水平均恢复到高于有效阈值(即435 ng/mL),只有1例患者在随访2时仍低于阈值(图1B)。与阿勒替尼单药治疗相比,西马鲁肽联合给药后,患者的毒性明显更大。胃肠道副作用,如呕吐(A期对B期:0%对80%)、恶心(A期对B期:0%对50%)和厌食(A期对B期:0%对50%)在给予西马鲁肽后更为普遍。根据不良事件通用术语标准(CTCAE v5.0),所有毒性均为1级或2级毒性,见补充表S4。回顾病人的日记,发现在服用西马鲁肽后有减少食物摄入量的趋势。由于这是第一个描述抗癌药物和西马鲁肽之间相互作用的研究,这些发现对临床医生和癌症患者具有重要意义。 如前所述,在回顾性分析的基础上建立了阿勒替尼的暴露-反应关系[b]。目前在一项前瞻性随机试验中研究了无进展生存期的阈值;即所谓的ADAPT-ALEC研究,该研究正在荷兰和法国招募患者(NCT05525338)。鉴于这种假定的暴露-反应关系,阿勒替尼和西马鲁肽联合使用可能会阻碍阿勒替尼的抗肿瘤疗效。在我们的研究中,10名受试者中有4名在西马鲁肽和阿勒替尼联合治疗后的谷底水平低于435 ng/mL阈值,而阿勒替尼单药治疗则没有。所有4例患者均服用阿勒替尼450 mg,每日两次(BID)。GLP-1受体激动剂的作用机制是多方面的。在某种程度上,对减肥的积极影响可能是由于通过模仿GLP-1诱导饱腹感,从而导致食欲下降。此外,GLP-1受体激动剂降低胃动力,导致摄入食物后的饱腹感延长。这种机制也会引起胃肠道的副作用,比如呕吐。在我们的试验中,恶心和呕吐导致5例患者至少漏服一次阿勒替尼,这可能导致阿勒替尼AUC0-10h的减少。尽管如此,大多数副作用是短暂的,仅发生在给药后的第一天(补充图S1)。值得注意的是,除了1例遗漏的阿勒替尼剂量外,所有患者都发生在B期的前两天,这表明在进行药代动力学采样时,所有患者的浓度都应该达到稳定状态。此外,事后敏感性分析发现遗漏剂量对阿勒替尼暴露没有影响(补充表S5)。相反,食物效应可能为观察到的西马鲁肽给药后阿勒替尼暴露减少提供了一个更合理的假设。值得注意的是,在我们的研究中,大多数患者由于食欲不振和胃肠道不良事件而难以坚持规定的饮食,这是众所周知的西马鲁肽的副作用。由于alectinib是口服给药,其吸收受到食物摄入[8]的显著影响,特别是即使在喂食条件下,生物利用度也很低(37%)。因此,食物摄入的改变将对全身alectinib暴露产生重大影响。在我们的研究中,由于食欲下降,大多数患者的食物(以及脂肪)摄入受损,可能会影响alectiinib的吸收。这一点尤其重要,因为另一项研究发现,服用西马鲁肽的患者报告说,他们对高脂肪食物的偏好减少了。在我们的试验中,西马鲁肽给药一次以评估药代动力学安全性。基于我们的结果,我们强调在开西马鲁肽处方时需要谨慎,特别是在阿勒替尼剂量减少的患者中。虽然对于阿勒替尼诱导的体重增加,semaglutide仍然是一种很有前景的治疗方法,但需要进一步的研究来评估semaglutide对阿勒替尼治疗的长期影响。例如,体重减轻本身与较低的alectinib清除率有关,这可能随后增加长期bbb的暴露。总之,我们的研究强调了一种临床相关的药代动力学药物与阿勒替尼之间的相互作用,导致阿勒替尼暴露量的显著减少,这很可能是由阿勒替尼介导的食物效应引起的。这强调了同时开具阿勒替尼和西马鲁肽(以及潜在的其他GLP-1受体激动剂)处方时谨慎操作的重要性,并且有必要监测这些患者的阿勒替尼血浆浓度。研究概念和设计:Niels Heersche, Daan A.C. Lanser, Stijn L.W. Koolen, Anne-Marie C. Dingemans, G.D. Marijn Veerman和Ron H.J. Mathijssen。研究表现:Niels Heersche, Daan A.C. Lanser, Attila Icli和Peter de Bruijn。患者选择:Marthe S. Paats和Anne-Marie C. Dingemans。数据分析和解释:Niels Heersche, Daan A.C. Lanser, Esther Oomen-de Hoop, Attila Icli, G.D. Marijn Veerman和Ron H.J. Mathijssen。原稿:Niels Heersche, Daan A.C. Lanser, G.D. Marijn Veerman和Ron H.J. Mathijssen。编辑:Niels Heersche, Daan A.C. Lanser, Esther Oomen-de Hoop, Attila Icli, Peter de Bruijn, Marthe S. Paats, Elisabeth F.C. van Rossum, Stijn L.W. Koolen, Ron H.N. van Schaik, Anne-Marie C. Dingemans, G.D. Marijn Veerman和Ron H.J. Mathijssen。EFCvR参与了Rhythm制药公司针对罕见遗传性肥胖的靶向治疗的临床试验(所有费用由该研究所支付)。ACD报告来自安进的资助(全部支付给研究所)。 RHJM报告了来自安斯泰来、拜耳、勃林格殷格翰、crystal Therapeutics、Deuter Oncology、Echo Pharmaceuticals、Nordic Pharma、诺华、Pamgene、辉瑞、罗氏、赛诺菲和施维雅的研究者发起的试验的无限制拨款(全部支付给该研究所)。其他作者均未报告coi。不适用。鹿特丹伊拉斯谟大学医学中心主管部门和当地伦理委员会批准了该试验(注册ID分别为NL78079.078.23和MEC 21-0478)。所有参与者在纳入研究之前都提供了书面知情同意书。荷兰审判登记处编号:NL9702
本文章由计算机程序翻译,如有差异,请以英文原文为准。
求助全文
约1分钟内获得全文 求助全文
来源期刊
Cancer Communications
Cancer Communications Biochemistry, Genetics and Molecular Biology-Cancer Research
CiteScore
25.50
自引率
4.30%
发文量
153
审稿时长
4 weeks
期刊介绍: Cancer Communications is an open access, peer-reviewed online journal that encompasses basic, clinical, and translational cancer research. The journal welcomes submissions concerning clinical trials, epidemiology, molecular and cellular biology, and genetics.
×
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