Skip to main content
Erschienen in: Journal of Translational Medicine 1/2022

Open Access 01.12.2022 | Research

Lenvatinib inhibits the growth of gastric cancer patient-derived xenografts generated from a heterogeneous population

verfasst von: John D. Karalis, Lynn Y. Yoon, Suntrea T. G. Hammer, Changjin Hong, Min Zhu, Ibrahim Nassour, Michelle R. Ju, Shu Xiao, Esther C. Castro-Dubon, Deepak Agrawal, Jorge Suarez, Scott I. Reznik, John C. Mansour, Patricio M. Polanco, Adam C. Yopp, Herbert J. Zeh III, Tae Hyun Hwang, Hao Zhu, Matthew R. Porembka, Sam C. Wang

Erschienen in: Journal of Translational Medicine | Ausgabe 1/2022

Abstract

Background

Lenvatinib is a multitargeted tyrosine kinase inhibitor that is being tested in combination with immune checkpoint inhibitors to treat advanced gastric cancer; however, little data exists regarding the efficacy of lenvatinib monotherapy. Patient-derived xenografts (PDX) are established by engrafting human tumors into immunodeficient mice. The generation of PDXs may be hampered by growth of lymphomas. In this study, we compared the use of mice with different degrees of immunodeficiency to establish PDXs from a diverse cohort of Western gastric cancer patients. We then tested the efficacy of lenvatinib in this system.

Methods

PDXs were established by implanting gastric cancer tissue into NOD.Cg-PrkdcscidIl2rgtm1Wjl/SzJ (NSG) or Foxn1nu (nude) mice. Tumors from multiple passages from each PDX line were compared histologically and transcriptomically. PDX-bearing mice were randomized to receive the drug delivery vehicle or lenvatinib. After 21 days, the percent tumor volume change (%Δvtumor) was calculated.

Results

23 PDX models were established from Black, non-Hispanic White, Hispanic, and Asian gastric cancer patients. The engraftment rate was 17% (23/139). Tumors implanted into NSG (16%; 18/115) and nude (21%; 5/24) mice had a similar engraftment rate. The rate of lymphoma formation in nude mice (0%; 0/24) was lower than in NSG mice (20%; 23/115; p < 0.05). PDXs derived using both strains maintained histologic and gene expression profiles across passages. Lenvatinib treatment (mean %Δvtumor: -33%) significantly reduced tumor growth as compared to vehicle treatment (mean %Δvtumor: 190%; p < 0.0001).

Conclusions

Nude mice are a superior platform than NSG mice for generating PDXs from gastric cancer patients. Lenvatinib showed promising antitumor activity in PDXs established from a diverse Western patient population and warrants further investigation in gastric cancer.
Hinweise

Supplementary Information

The online version contains supplementary material available at https://​doi.​org/​10.​1186/​s12967-022-03317-7.
John D. Karalis and Lynn Y. Yoon contributed equally as primary authors. Matthew R. Porembka and Sam C. Wang contributed equally as co-corresponding authors.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Introduction

Gastric cancer is the third-leading cause of cancer-related deaths worldwide and was responsible for an estimated 769,000 deaths in 2020 [1]. The overall 5-year survival rate for gastric cancer patients is only 32% in the United States, and novel therapies are urgently needed [2]. However, efforts to develop effective anticancer therapies have been impeded by a lack of preclinical models that recapitulate the heterogeneity of gastric tumors. One strategy to overcome this limitation is the generation of patient-derived xenografts (PDX), in which human tumors are propagated in immunodeficient mice. PDXs are a completely in vivo preclinical platform that bypasses growing the tumor cells in in vitro environments under highly artificial conditions. As a result, PDXs retain a high degree of the histologic, genomic, and transcriptomic characteristics of the human tumors from which they are derived [35]. PDXs also exhibit superior predictive values for clinical outcomes than traditional in vitro cell culture and cell line-derived xenografts [6]. While gastric cancer PDX models have been established by East Asian and European institutions, few PDXs have been generated from Black/African American or Hispanic patients [4, 7, 8]. This is a significant deficiency because gastric cancer biology, and response to novel therapies, has been shown to vary by patient race and ethnicity [914].
The strain of recipient mice that should be used to establish gastric cancer PDX has not been determined. Commonly used strains include NOD.Cg-Prkdcscid Il2rgtm1Wjl/SzJ mice (NSG) and Foxn1nu (nude) mice [5, 15]. NSG mice are highly immunodeficient and lack mature T cells, B cells, and natural killer cells and have defective macrophages and dendritic cells, whereas nude mice only lack mature T cells [16]. In theory, highly immunosuppressed strains may allow for higher tumor engraftment rates. However, the use of immunodeficient mice to generate solid tumor PDXs is hampered by the growth of B cell lymphoma, the frequency of which may be affected by the degree of immunodeficiency of the recipient mice [4, 5].
Lenvatinib is a multitargeted tyrosine kinase inhibitor and inhibits vascular endothelial growth factor receptors 1–3 (VEGFR 1–3), fibroblast growth factor receptors 1–4 (FGFR 1–4), platelet-derived growth factor receptors α and β (PDGFR α-β), RET (rearranged during transfection), and c-kit. However, it most potently affects the VEGFR family, suggesting a dominant anti-angiogenic mechanism [17, 18]. Anti-VEGFR therapy does have a role in gastric cancer treatment; ramicirumab, which is an anti-VEGFR2 monoclonal antibody, is approved for second-line therapy in advanced gastric cancer [19]. Recently, lenvatinib showed promising efficacy when given in combination with pembrolizumab, an immune checkpoint inhibitor, for the treatment of patients with advanced gastric cancer in a phase II clinical trial in Japan [20]. However, lenvatinib has not been well studied in the gastric cancer context as a monotherapy [21]. Thus, additional preclinical and clinical data are needed to understand the effects of lenvatinib monotherapy in a diverse gastric cancer patient population.
In this study, we generated PDXs from Western gastric cancer patients of various racial and ethnic backgrounds. We used NSG or nude mice as recipients to perform a head-to-head comparison of their rates of engraftment and lymphomatous growth. Finally, we used our PDX model to evaluate the efficacy of lenvatinib.

Materials and methods

Sample acquisition

This study was approved by the University of Texas Southwestern Medical Center institutional review board. All patients with gastric adenocarcinoma who were treated at Parkland Memorial Hospital (Dallas, TX) and William P. Clements Jr. University Hospital (Dallas, TX) were approached for the study. All enrolled patients provided written informed consent.
Tumor tissue was obtained from patients by endoscopic biopsy, gastric resection, and biopsy of metastatic sites. Samples for PDX generation were immediately placed into sterile phosphate buffered saline and transported on ice to the mouse procedure facility for implantation. Samples for transcriptomic analysis were immediately placed in RNAlater (Invitrogen) for at least 24 h at 4 °C and then transferred to − 80 °C.

Mice

All mice were handled in accordance with the guidelines of the institutional animal care and use committee at the University of Texas Southwestern Medical Center. NSG and nude mice were obtained from Jackson Laboratory.

PDX generation, passage, storage, and reanimation

Samples obtained by esophagogastroduodenoscopy (EGD) biopsy (each 1–3 mm3), biopsy of metastatic sites (1–3 mm3), and surgical resection (3–5 mm3) were coated in Matrigel Matrix (Corning Life Sciences) and implanted into the subcutaneous space in the flanks of recipient mice. Tumors were passaged when they reached a maximum diameter of 1.5 cm or skin necrosis overlying the tumor was observed. We considered a PDX line to be established if the tumor grew to a diameter of at least 1.5 cm and grew after passage to a second recipient mouse. At passage, the tumor was excised from the subcutaneous tissue, divided, and then: (1) passaged into the flanks of a new mouse, (2) fixed in 4% paraformaldehyde for histologic evaluation, and (3) stored in 10% dimethyl sulfoxide (DMSO) + 90% fetal bovine serum and submerged in liquid nitrogen for future use. To reanimate frozen PDX tissue, the tissue was warmed in a 37 °C water bath for 60 s and then implanted in an identical manner as a fresh sample. After reanimation, mice were maintained the same way as mice with primarily implanted xenografts.

Lenvatinib testing

Four PDX lines were selected for drug testing. When tumors reached a volume of 300–500 mm3, mice were randomized to receive either the drug delivery vehicle (DMSO mixed with corn oil) or lenvatinib (MedChemExpress; cat #HY-10981) 10 mg/kg daily by oral gavage. Tumor dimensions were measured at baseline and once weekly thereafter by a blinded observer. After 21 days, all mice were euthanized. Tumor volume was calculated using the formula (π/6) × length × width2.[22] The percent tumor volume change (%Δvtumor) from baseline of all tumors was calculated. Mice were weighed weekly.

Histology, immunohistochemistry, and immunofluorescence

Tissue samples were fixed in 4% paraformaldehyde for 24 h and then embedded in paraffin. Sections were cut to 4 μm. All samples were stained with hematoxylin and eosin (H&E) for histologic examination. Tumor and PDX histology were evaluated in a blinded fashion by a board-certified pathologist with expertise in gastrointestinal malignancies (S.T.G.H.).
Antibodies used for immunohistochemistry (IHC) were mouse anti-human pan-cytokeratin AE1/AE3 monoclonal antibody (1:500 dilution; Santa Cruz Biotechnology; SC-81714), mouse anti-human CD20 monoclonal antibody (Agilent; IR60461-2), rabbit anti-human CD31 monoclonal antibody (1:50 dilution; Abcam; ab28364), rabbit anti-human Ki-67 antibody (1:1000 dilution; Abcam, ab15580). Terminal deoxynucleotidyl transfer dUTP nick end label (TUNEL) immunofluorescence (IF) was performed using the In Situ Cell Death Detection Kit, AP (Roche). Hoechst 33342, Trihydrochloride, Trihydrate (Invitrogen) was used for nuclear counterstaining.

Quantification of intratumoral vascular density, tumor cell proliferation, and apoptosis

Blood vessel density was quantified using the hot spot method [23]. In brief, each slide was scanned under light microscopy at 40× magnification to identify the three areas of highest vascular density. Those three areas were then evaluated at 200× magnification and the number of vessels per 200× magnification field were counted. Any stained ring of endothelial cells surrounding a lumen or discrete cluster of endothelial cells was counted as a single vessel. Two tumors from each PDX line were evaluated. Ki-67-positive cells were manually counted in three 400× fields per tumor. The Ki-67 of two tumors per treatment arm was calculated, and their average value was reported. To quantify TUNEL-positive cells, each slide was scanned under fluorescent microscopy at 40× magnification to identify the three areas of highest TUNEL-positivity. Those three areas were then evaluated at 200× magnification and the number of apoptotic cells per 200× magnification field were manually counted. The TUNEL-positivity rate of two tumors per treatment arm was calculated, and their average value was reported. All quantification was performed in a blinded fashion.

RNA sequencing and analysis

RNA was extracted using the RNeasy Mini Kit (Qiagen). Total RNA was quantified using the Ribogreen method using Victor X2 fluorometry (Life Technologies). RNA integrity was assessed using an Agilent Technologies 2100 TapeStation. Libraries were made with TruSeq Stranded Total RNA kits (Illumina) and sequenced on a NovaSeq 6000 with 2 × 150 base pair paired-end reads.
RNA-sequencing data was refined by Fast p v0.20.1 (default settings) such that Illumina universal adapters were trimmed and reads with lower quality bases, or noninformative reads, were filtered out. A total of 51.3 million paired-end reads were retained. All samples engrafted into mice were aligned into both human (hg19) and mouse reference assembly (m38) using STAR (v2.7.9, —twopassMode Basic). Each aligned read was examined to determine the organism of origin. Reads uniquely aligned to the mouse genome (i.e., with a higher matching score than the human genome) were removed. Then, the refined human reads were converted to transcriptome abundant matrix in the format of transcripts per kilobase million (TPM) via Salmon v1.5.2 with input parameters: ‘-I IU—validateMappings’). The Salmon reference genome sequence index was built on human reference transcript sequences (GRCm38.v29) with the gene model Gencode v29 (Ensembl 94).

Statistical analysis

Categorical variables were presented as counts and compared with chi-square or Fisher’s exact tests. The %Δvtumor between vehicle and lenvatinib-treated tumors was compared with Mann–Whitney U tests, and the same tests were used for comparisons between vehicle and lenvatinib-treated vessel counts. A p value of < 0.05 was considered statistically significant. Statistical computation was performed using GraphPad Prism (Version 9.1.0).

Results

Gastric cancer PDXs were generated from a diverse Western population

Tumor samples were collected from 139 gastric cancer patients from 2015 to 2020. The baseline characteristics of the patients are summarized in Table 1. The median patient age was 55 years (range: 25–89 years), and 65% of patients were male. 55% of samples were obtained from patients who self-identified as being of Hispanic origin, 19% were from Black/African American patients, 4% were from Asian patients, 1% was from an American Indian patient, and 22% were from non-Hispanic White patients. 56% of patients presented with locally advanced tumors (T2-4N0M0, TanyN1-3M0) and 42% of patients presented with metastatic disease (TanyNanyM1). Only 2% of patients presented with local disease (T1-2N0M0). 46% of patient tumors were of Lauren diffuse-type histology, 40% were intestinal-type, and 14% were mixed-type.
Table 1
Patient demographic, clinical, and pathologic factors
Variable
n (%)
Gender
 Male
91 (65)
 Female
48 (35)
Race/ethnicity
 Non-Hispanic White
30 (22)
 Black
26 (19)
 Hispanic
77 (55)
 Asian
5 (4)
 American Indian
1 (1)
Presenting stage
 Local
2 (2)
 Locally advanced
78 (56)
 Metastatic
59 (42)
Lauren classification
 Intestinal
47 (40)
 Diffuse
54 (46)
 Mixed
17 (14)
 Unknown
21
Differentiation
 Well
1 (1)
 Moderate
31 (23)
 Poor
102 (76)
 Unknown
5
H. pylori
 Negative
82 (73)
 Positive
30 (27)
 Unknown
27
HER2
 0–1+ or 2+ NAmp
72 (77)
 3+ or 2+ & amplified
21 (23)
 Unknown
46
A HER2 score of 3+ or 2+ and fluorescent in situ hybridization (FISH) amplified was considered positive. A HER2 score of 0–1+ or 2+ and FISH non-amplified was considered negative
Amp, fluorescence in situ hybridization (FISH) amplified; Namp, FISH non-amplified
Overall, samples from 17% (23/139) of patients successfully engrafted (Table 2). 10 PDX lines were generated from Black/African American patients, 9 from Hispanic patients, 3 from non-Hispanic White patients, and 1 from an Asian patient. Next, we investigated whether clinicopathologic characteristics of the patient tumor were associated with PDX engraftment. We found that tumors from Black patients engrafted at a higher rate than other racial/ethnic groups and that Lauren intestinal-type tumors engrafted at a higher rate than diffuse-type tumors. Engraftment rate was not associated with whether the sample was obtained from EGD, surgical resection, or biopsy of a metastatic site (Table 2).
Table 2
Patient demographic, clinical, and pathologic factors associated with PDX engraftment
Variable
n (%)
P value
Engrafted (n = 23)
Not engrafted (n = 116)
Gender
 Male
19 (83)
72 (62)
NS
 Female
4 (17)
44 (38)
Race/ethnicity
 Non-Hispanic White
3 (13)
27 (23)
< 0.05
 Black/African American
10 (43)
16 (14)
 Hispanic
9 (39)
68 (59)
 Asian
1 (4)
4 (3)
 American Indian
0 (0)
1 (1)
Presenting stage
 Local
0 (0)
2 (2)
NS
 Locally advanced
13 (57)
65 (56)
 Metastatic
10 (43)
49 (42)
Lauren classification
 Intestinal
14 (61)
33 (35)
< 0.01
 Diffuse
3 (13)
51 (54)
 Mixed
6 (26)
11 (12)
 Unknown
0
21
Differentiation
 Well
0 (0)
1 (1)
NS
 Moderate
7 (30)
24 (22)
 Poor
16 (70)
86 (77)
 Unknown
0
5
H. pylori infection
 Negative
15 (79)
67 (72)
NS
 Positive
4 (21)
26 (28)
 Unknown
4
23
HER2 status
 0–1+ or 2+ NAmp
12 (75)
60 (78)
NS
 3+ or 2+ Amp
4 (25)
17 (22)
 Unknown
7
39
Tissue sourcea
 EGD biopsy
9 (39)
96 (54)
NS
 Resection
8 (35)
56 (32)
 Metastasis biopsy
6 (26)
25 (14)
A HER2 score of 3+ or 2+ and fluorescent in situ hybridization (FISH) amplified was considered positive. A HER2 score of 0–1+ or 2+ and FISH non-amplified was considered negative
Amp, fluorescence in situ hybridization (FISH) amplified; EGD, esophagogastroduodenoscopy; NAmp, FISH non-amplified
aMultiple samples were collected from some patients

Gastric cancer PDXs are histologically and transcriptomically similar to the parent tumors from which they were derived

Engrafted PDXs retained the histologic characteristics of the human primary tumors they were derived from, including after the PDX was passaged, regardless of whether the PDX was propagated in NSG or nude mice (Fig. 1). We also compared the transcriptomic profiles of the tumors via bulk RNA sequencing. We first analyzed the similarity of gene expression between the human primary tumor and an early passage PDX. We found a modest correlation (Pearson correlation coefficient 0.45–0.69, Fig. 2); however, this was expected because the human primary tumor sample also included cells of the human tumor microenvironment such as fibroblasts and immune cells; whereas the PDX tumor, due to its growth in an immunodeficient mouse, has fewer infiltrating immune cells. We then compared an early passage PDX with a subsequently passaged PDX and found a strong correlation between their gene expression profiles (Pearson correlation coefficient 0.68–0.88, Fig. 2) which indicates that our PDX model exhibits a high degree of transcriptomic stability, after multiple passages. Notably, we found similar stability of gene expression between PDXs propagated in NSG and nude mice.
The median time to initial PDX engraftment was 13.3 weeks while the median number of weeks between subsequent passages was significantly lower at 6.8 weeks (p < 0.0001; Fig. 3A, B). There was no difference in time to engraftment based on whether the tissue was obtained by EGD, surgical resection, or biopsy of a metastatic site (Fig. 3C). All PDXs stored in liquid nitrogen were able to be successfully reanimated, even after more than 5 years.

Nude mice are a better recipient than NSG mice for PDX generation

We first tested NSG mice as recipients for PDXs. We found that while 36% (41/115) of samples resulted in tumor growth, 56% (23/41) of the tumors were lymphomas, as confirmed by H&E staining, negative pan-cytokeratin AE1/AE3 IHC, and positive CD20 IHC. The remaining 18 tumors were histologically consistent with gastric adenocarcinoma and were pan-cytokeratin AE1/AE3-positive, and CD20-negative. Thus, the rate of successful PDX generation in NSG mice was 16% (18/115). We then tested nude mice as the recipient platform and found a similarly successful PDX establishment rate of 21% (5/24; Table 3). There was no difference in median time to initial gastric cancer engraftment between the strains (NSG: 12.8 weeks, nude: 19.1 weeks; p = NS). Importantly, we observed no lymphomas in nude mice. The difference in lymphoma formation in NSG (20%; 23/115) and nude mice (0%; 0/24) was significant (Table 3; p < 0.05).
Table 3
Comparison of NSG and nude mice as recipients for gastric cancer PDX generation
 
n (%)
P value
NSG (n = 115)
Nude (n = 24)
Gastric adenocarcinoma
18 (16)
5 (21)
NS
Lymphoma
23 (20)
0 (0)
< 0.05

Transplant of a mixed gastric cancer-lymphoma PDX from an NSG to a nude mouse eradicated lymphoma

One sample implanted into an NSG mouse resulted in a tumor that contained both gastric adenocarcinoma and lymphoma in an approximate 1:1 ratio (Fig. 4A). The presence of a mixed population was confirmed with pan-cytokeratin and CD20 IHC (Fig. 4B, C). Since we had previously found that no lymphomas developed in nude mice, we hypothesized that we could “rescue” the gastric adenocarcinoma component by propagating the mixed tumor in nude mice. We passaged the tumor into nude mice and analyzed the tumor that formed. We found that the tumor histology was consistent with gastric adenocarcinoma and no lymphoma was observed (Fig. 4D). All tumor cells were pan-cytokeratin positive (Fig. 4E) and there were no CD20-positive cells (Fig. 4F). These histological findings were maintained on subsequent passages. Overall, these data support the notion that the lymphomatous component was eradicated upon passaging in nude mice.

Lenvatinib inhibited the growth of gastric cancer PDXs and reduced intratumoral vascular density

We then evaluated the utility of our PDX model for preclinical drug testing. Four PDX lines with heterogeneous features were selected for testing. These PDX lines were chosen to represent patients from multiple racial/ethnic groups, Lauren diffuse- and intestinal-type tumors, chemotherapy-treated and chemotherapy-naïve tumors, as well as both gastric and gastroesophageal junction tumors (Additional file 1: Table S1). Lenvatinib inhibited the growth of all four unique PDX lines (%Δvtumor range: − 2% to − 63%). An analysis of all vehicle (n = 11) and lenvatinib-treated (n = 11) PDXs yielded a mean %Δvtumor of 190% and − 33%, respectively (p < 0.0001; Fig. 5A–C). Vehicle-treated mice had an average change in body weight of 0.2 g (− 1%) and lenvatinib-treated mice had an average change of − 1.4 g (− 6.0%; p < 0.05; Fig. 5D). No unscheduled euthanasia was required.
H&E examination of vehicle and lenvatinib-treated tumors revealed markedly increased tumor necrosis and desmoplasia in the lenvatinib-treated samples (Fig. 6A). Because the dominant action of lenvatinib is VEGF receptor inhibition, which would impede tumor angiogenesis, we next performed IHC directed at the endothelial cell marker CD31. The average number of vessels per 200 × field in the vehicle group was 12.4 (range: 7–16) versus 1.6 (range: 0–2.5) in the lenvatinib-treated group (p < 0.05; Fig. 6A, B), thus confirming that lenvatinib therapy had the on-target effect of reducing tumor vasculature. We next determined the effect of lenvatinib treatment on tumor cell proliferation and apoptosis. We found no difference in Ki-67 percentage between vehicle and lenvatinib-treated tumors (Fig. 6C, D). However, we did find a significantly increased number of apoptotic cells in lenvatinib-treated tumors (Fig. 6E, F).

Discussion

In this study, we established 23 gastric cancer PDX lines from patients of diverse racial and ethnic backgrounds. PDXs are unique among preclinical models in that they recapitulate the native tumor biology of the patients from whom they are derived. However, to date, gastric cancer PDXs have primarily been generated from European and East Asian patients [4, 7, 8]. The lack of diversity in previously generated gastric cancer PDXs is a major shortcoming for the field as there is significant biological heterogeneity associated with patient race and ethnicity [914]. For example, Strong et al. [9] found that patients in Korea had improved gastric cancer-specific survival when compared to patients in the US, even after controlling for other prognostic variables. Additionally, differing races and ethnicities have exhibited divergent responses to therapy; for example, in a phase II study of regorafenib, Asian patients were found to have a more favorable response than patients of other races and ethnicities [11]. To our knowledge, this is the first description of PDXs generated from a large number of Black/African American and Hispanic patients.
The preferred recipient mouse strain for PDX generation has not been established [4, 5]. Theoretically, a highly immunodeficient mouse strain, such as the NSG line that lacks mature T cells, B cells, and natural killer cells, may allow for a higher tumor engraftment rate, but at a cost of increased lymphomatous growth as compared to a modestly immunodeficient strain, such as nude mice, which lack only mature T cells [16]. To answer this question, we performed a head-to-head comparison of NSG and nude mice as recipients. We found that while engraftment rates were similar between the two strains, there was a lower rate of lymphoma formation in nude mice. The unregulated proliferation of xenografted patient B cells leads to lymphoma formation in immunodeficient mice. Our findings demonstrate that the presence of some functional elements of the immune system in nude mice is sufficient to suppress lymphoma overgrowth without affecting the rate of gastric adenocarcinoma engraftment. Moreover, we found that PDXs engrafted into NSG and nude mice similarly retained the histologic and transcriptomic characteristics of the human primary tumor from which they were derived. While administration of the anti-CD20 antibody rituximab has been suggested as a method to reduce lymphomatous transformation in gastric cancer PDXs, we have not found this to be necessary due to the success of our nude mouse protocol [15, 24]. Thus, our results demonstrate that nude mice are a preferred platform over NSG mice for gastric cancer PDX generation. Our findings are consistent with the report by Choi et al. who also concluded that nude mice are the preferred recipient for gastric cancer PDX generation [5]. This has significant logistical implications as nude mice are less expensive than NSG mice and may have fewer housing and handling restrictions.
The efficacy of novel therapies in the preclinical setting is infrequently replicated in clinical trials [25]. This may be explained, in part, by the degree to which preclinical research relies on cancer cell lines that are established as a monolayer growing in a nutrient-rich environment. In contrast, PDXs are unmodified by in vitro adaptation and have been shown to maintain a high degree of the histologic, genomic, and transcriptomic features of the parent tumors from which they were derived [5, 26]. As a result, PDXs are a demonstrably better platform for preclinical drug testing [27].
We found that lenvatinib inhibited the growth of all four PDX lines tested, regardless of patient race/ethnicity, Lauren subtype, histologic differentiation, anatomic tumor location, and history of prior chemotherapy treatment. This suggests that lenvatinib monotherapy may be effective against a broad range of gastric cancers, although there is a paucity of data in gastric cancer patients. Lenvatinib has only been tested as a monotherapy in 6 advanced gastric cancer patients in a phase I study in which 3 of the 6 patients showed stable disease [21]. In the KEYNOTE-061 trial, the objective response rate of gastric cancer patients treated with pembrolizumab was approximately 15% in patients with a combined positive score (CPS) of ≥ 1 [28]. In contrast, Kawazoe et al.’s findings from a phase II trial that tested a combination of lenvatinib plus pembrolizumab showed an objective response in 69% of patients, despite only 66% of the patient cohort having a CPS of ≥ 1 [20]. As such, we reasoned that in the Kawazoe et al. cohort, either: (1) lenvatinib was the primary driver of the increased response rates observed, or (2) lenvatinib enhanced the efficacy of pembrolizumab. Because we found that lenvatinib was effective in the absence of concurrent immune checkpoint inhibition, our data suggest that lenvatinib may, at least in part, be an independent driver of the results observed in the Kawazoe et al. study. Another notable finding was the efficacy of lenvatinib in all three treatment-naïve PDX lines. This is significant because ramucirumab, an anti-angiogenic monoclonal antibody that blocks VEGFR-2, has demonstrated efficacy only in a second-line setting [29]. The broader range of lenvatinib’s anti-angiogenic inhibition via blockade of VEGFR1-3 may be more effective for the treatment of gastric cancer than selective VEGFR-2 inhibition that ramucirumab provides.
There are several limitations to our study. There is interest in using PDXs as a “patient avatar” to test therapies and identify the treatment options on an individualized basis. However, we were only able to generate PDXs from approximately 1 in 6 patients and our median time to engraftment was 13 weeks. Therefore, while this personalized approach is attractive in principle, it may not be feasible at most centers. Additionally, PDX-bearing mice are immunocompromised; as such, tumor-immune interactions and immune-oncologic therapies cannot be easily evaluated in PDXs. Finally, the influence of the tumor microenvironment in the subcutaneous space of the mouse flank is different than influences from gastric tissue and the unique environment of the stomach lumen. Despite these limitations, we believe that PDXs are a robust model for generating high-quality preclinical data for novel gastric cancer therapies.

Conclusion

In this report, we describe the successful establishment of a PDX model from a diverse gastric cancer patient population. We recommend nude mice as the preferred recipient strain over NSG mice for gastric cancer PDX generation as we found lymphomas were unable to grow in nude mice. Finally, lenvatinib is a promising agent for the treatment of gastric cancer and warrants further investigation in clinical trials beyond in combination with pembrolizumab.

Acknowledgements

We acknowledge the assistance of the Tissue Management Shared Resource at the Simmons Comprehensive Cancer Center at the University of Texas Southwestern Medical Center, which is supported in part by the National Cancer Institute under award number P30 CA142543. Figure 4 was created with BioRender.com. The authors would like to thank Dave Primm of the UT Southwestern Department of Surgery for help in editing this article.

Declarations

This study was approved by the University of Texas Southwestern Medical Center institutional review board.
Not applicable.

Competing interests

The authors declare that they have no competing interests.
Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Literatur
4.
Zurück zum Zitat Corso S, et al. A comprehensive PDX gastric cancer collection captures cancer cell-intrinsic transcriptional MSI traits. Can Res. 2019;79(22):5884–96.CrossRef Corso S, et al. A comprehensive PDX gastric cancer collection captures cancer cell-intrinsic transcriptional MSI traits. Can Res. 2019;79(22):5884–96.CrossRef
5.
Zurück zum Zitat Choi YY, et al. Establishment and characterisation of patient-derived xenografts as paraclinical models for gastric cancer. Sci Rep. 2016;6(1):1–12.CrossRef Choi YY, et al. Establishment and characterisation of patient-derived xenografts as paraclinical models for gastric cancer. Sci Rep. 2016;6(1):1–12.CrossRef
6.
Zurück zum Zitat Evrard YA, et al. Systematic establishment of robustness and standards in patient-derived xenograft experiments and analysis. Can Res. 2020;80(11):2286–97.CrossRef Evrard YA, et al. Systematic establishment of robustness and standards in patient-derived xenograft experiments and analysis. Can Res. 2020;80(11):2286–97.CrossRef
8.
Zurück zum Zitat Zhu Y, et al. Establishment and characterization of patient-derived tumor xenograft using gastroscopic biopsies in gastric cancer. Sci Rep. 2015;5(1):8542.CrossRef Zhu Y, et al. Establishment and characterization of patient-derived tumor xenograft using gastroscopic biopsies in gastric cancer. Sci Rep. 2015;5(1):8542.CrossRef
9.
Zurück zum Zitat Strong VE, et al. Comparison of gastric cancer survival following R0 resection in the United States and Korea using an internationally validated nomogram. Ann Surg. 2010;251(4):640–6.CrossRef Strong VE, et al. Comparison of gastric cancer survival following R0 resection in the United States and Korea using an internationally validated nomogram. Ann Surg. 2010;251(4):640–6.CrossRef
10.
Zurück zum Zitat Tan D, Mok T, Rebbeck TR. Cancer genomics: diversity and disparity across ethnicity and geography. J Clin Oncol. 2016;34(1):91–101.CrossRef Tan D, Mok T, Rebbeck TR. Cancer genomics: diversity and disparity across ethnicity and geography. J Clin Oncol. 2016;34(1):91–101.CrossRef
11.
Zurück zum Zitat Pavlakis N, et al. Regorafenib for the treatment of advanced gastric cancer (INTEGRATE): a multinational placebo-controlled phase II trial. J Clin Oncol. 2016;34(23):2728–35.CrossRef Pavlakis N, et al. Regorafenib for the treatment of advanced gastric cancer (INTEGRATE): a multinational placebo-controlled phase II trial. J Clin Oncol. 2016;34(23):2728–35.CrossRef
12.
Zurück zum Zitat Sanjeevaiah A, et al. Gastric cancer: recent molecular classification advances, racial disparity, and management implications. J Oncol Pract. 2018;14(4):217–24.CrossRef Sanjeevaiah A, et al. Gastric cancer: recent molecular classification advances, racial disparity, and management implications. J Oncol Pract. 2018;14(4):217–24.CrossRef
13.
Zurück zum Zitat Chandra R, et al. The changing face of gastric cancer: epidemiologic trends and advances in novel therapies. Cancer Gene Ther. 2021;28(5):390–9.CrossRef Chandra R, et al. The changing face of gastric cancer: epidemiologic trends and advances in novel therapies. Cancer Gene Ther. 2021;28(5):390–9.CrossRef
14.
Zurück zum Zitat Wang SC, et al. Hispanic/Latino patients with gastric adenocarcinoma have distinct molecular profiles including a high rate of germline CDH1 variants. Can Res. 2020;80(11):2114–24.CrossRef Wang SC, et al. Hispanic/Latino patients with gastric adenocarcinoma have distinct molecular profiles including a high rate of germline CDH1 variants. Can Res. 2020;80(11):2114–24.CrossRef
15.
Zurück zum Zitat Corso S, et al. Rituximab treatment prevents lymphoma onset in gastric cancer patient-derived xenografts. Neoplasia. 2018;20(5):443–55.CrossRef Corso S, et al. Rituximab treatment prevents lymphoma onset in gastric cancer patient-derived xenografts. Neoplasia. 2018;20(5):443–55.CrossRef
16.
Zurück zum Zitat Brian W, Soper P. Modeling human cancer in mice. The Jackson Laboratory. Brian W, Soper P. Modeling human cancer in mice. The Jackson Laboratory.
17.
Zurück zum Zitat Cabanillas ME, Habra MA. Lenvatinib: role in thyroid cancer and other solid tumors. Cancer Treat Rev. 2016;42:47–55.CrossRef Cabanillas ME, Habra MA. Lenvatinib: role in thyroid cancer and other solid tumors. Cancer Treat Rev. 2016;42:47–55.CrossRef
18.
Zurück zum Zitat Matsui J, et al. E7080, a novel inhibitor that targets multiple kinases, has potent antitumor activities against stem cell factor producing human small cell lung cancer H146, based on angiogenesis inhibition. Int J Cancer. 2008;122(3):664–71.CrossRef Matsui J, et al. E7080, a novel inhibitor that targets multiple kinases, has potent antitumor activities against stem cell factor producing human small cell lung cancer H146, based on angiogenesis inhibition. Int J Cancer. 2008;122(3):664–71.CrossRef
19.
Zurück zum Zitat Fuchs CS, et al. Ramucirumab monotherapy for previously treated advanced gastric or gastro-oesophageal junction adenocarcinoma (REGARD): an international, randomised, multicentre, placebo-controlled, phase 3 trial. Lancet. 2014;383(9911):31–9.CrossRef Fuchs CS, et al. Ramucirumab monotherapy for previously treated advanced gastric or gastro-oesophageal junction adenocarcinoma (REGARD): an international, randomised, multicentre, placebo-controlled, phase 3 trial. Lancet. 2014;383(9911):31–9.CrossRef
20.
Zurück zum Zitat Kawazoe A, et al. Lenvatinib plus pembrolizumab in patients with advanced gastric cancer in the first-line or second-line setting (EPOC1706): an open-label, single-arm, phase 2 trial. Lancet Oncol. 2020;21(8):1057–65.CrossRef Kawazoe A, et al. Lenvatinib plus pembrolizumab in patients with advanced gastric cancer in the first-line or second-line setting (EPOC1706): an open-label, single-arm, phase 2 trial. Lancet Oncol. 2020;21(8):1057–65.CrossRef
21.
Zurück zum Zitat Boss DS, et al. A phase I study of E7080, a multitargeted tyrosine kinase inhibitor, in patients with advanced solid tumours. Br J Cancer. 2012;106(10):1598–604.CrossRef Boss DS, et al. A phase I study of E7080, a multitargeted tyrosine kinase inhibitor, in patients with advanced solid tumours. Br J Cancer. 2012;106(10):1598–604.CrossRef
22.
Zurück zum Zitat Shaw R, et al. Design, analysis and reporting of tumor models. Lab Anim. 2017;46(5):207–11.CrossRef Shaw R, et al. Design, analysis and reporting of tumor models. Lab Anim. 2017;46(5):207–11.CrossRef
23.
Zurück zum Zitat Weidner N, et al. Tumor angiogenesis and metastasis—correlation in invasive breast carcinoma. N Engl J Med. 1991;324(1):1–8.CrossRef Weidner N, et al. Tumor angiogenesis and metastasis—correlation in invasive breast carcinoma. N Engl J Med. 1991;324(1):1–8.CrossRef
24.
Zurück zum Zitat Fujii E, et al. Characterization of EBV-related lymphoproliferative lesions arising in donor lymphocytes of transplanted human tumor tissues in the NOG mouse. Exp Anim. 2014;63(3):289–96.CrossRef Fujii E, et al. Characterization of EBV-related lymphoproliferative lesions arising in donor lymphocytes of transplanted human tumor tissues in the NOG mouse. Exp Anim. 2014;63(3):289–96.CrossRef
25.
Zurück zum Zitat Johnson J, et al. Relationships between drug activity in NCI preclinical in vitro and in vivo models and early clinical trials. Br J Cancer. 2001;84(10):1424–31.CrossRef Johnson J, et al. Relationships between drug activity in NCI preclinical in vitro and in vivo models and early clinical trials. Br J Cancer. 2001;84(10):1424–31.CrossRef
26.
Zurück zum Zitat Zhu M, et al. Uncovering biological factors that regulate hepatocellular carcinoma growth using patient-derived xenograft assays. Hepatology. 2020;72(3):1085–101.CrossRef Zhu M, et al. Uncovering biological factors that regulate hepatocellular carcinoma growth using patient-derived xenograft assays. Hepatology. 2020;72(3):1085–101.CrossRef
27.
Zurück zum Zitat Gao H, et al. High-throughput screening using patient-derived tumor xenografts to predict clinical trial drug response. Nat Med. 2015;21(11):1318–25.CrossRef Gao H, et al. High-throughput screening using patient-derived tumor xenografts to predict clinical trial drug response. Nat Med. 2015;21(11):1318–25.CrossRef
28.
Zurück zum Zitat Shitara K, et al. Pembrolizumab versus paclitaxel for previously treated, advanced gastric or gastro-oesophageal junction cancer (KEYNOTE-061): a randomised, open-label, controlled, phase 3 trial. Lancet. 2018;392(10142):123–33.CrossRef Shitara K, et al. Pembrolizumab versus paclitaxel for previously treated, advanced gastric or gastro-oesophageal junction cancer (KEYNOTE-061): a randomised, open-label, controlled, phase 3 trial. Lancet. 2018;392(10142):123–33.CrossRef
29.
Zurück zum Zitat Wilke H, et al. Ramucirumab plus paclitaxel versus placebo plus paclitaxel in patients with previously treated advanced gastric or gastro-oesophageal junction adenocarcinoma (RAINBOW): a double-blind, randomised phase 3 trial. Lancet Oncol. 2014;15(11):1224–35.CrossRef Wilke H, et al. Ramucirumab plus paclitaxel versus placebo plus paclitaxel in patients with previously treated advanced gastric or gastro-oesophageal junction adenocarcinoma (RAINBOW): a double-blind, randomised phase 3 trial. Lancet Oncol. 2014;15(11):1224–35.CrossRef
Metadaten
Titel
Lenvatinib inhibits the growth of gastric cancer patient-derived xenografts generated from a heterogeneous population
verfasst von
John D. Karalis
Lynn Y. Yoon
Suntrea T. G. Hammer
Changjin Hong
Min Zhu
Ibrahim Nassour
Michelle R. Ju
Shu Xiao
Esther C. Castro-Dubon
Deepak Agrawal
Jorge Suarez
Scott I. Reznik
John C. Mansour
Patricio M. Polanco
Adam C. Yopp
Herbert J. Zeh III
Tae Hyun Hwang
Hao Zhu
Matthew R. Porembka
Sam C. Wang
Publikationsdatum
01.12.2022
Verlag
BioMed Central
Erschienen in
Journal of Translational Medicine / Ausgabe 1/2022
Elektronische ISSN: 1479-5876
DOI
https://doi.org/10.1186/s12967-022-03317-7

Weitere Artikel der Ausgabe 1/2022

Journal of Translational Medicine 1/2022 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Die „Zehn Gebote“ des Endokarditis-Managements

30.04.2024 Endokarditis Leitlinie kompakt

Worauf kommt es beim Management von Personen mit infektiöser Endokarditis an? Eine Kardiologin und ein Kardiologe fassen die zehn wichtigsten Punkte der neuen ESC-Leitlinie zusammen.

Strenge Blutdruckeinstellung lohnt auch im Alter noch

30.04.2024 Arterielle Hypertonie Nachrichten

Ältere Frauen, die von chronischen Erkrankungen weitgehend verschont sind, haben offenbar die besten Chancen, ihren 90. Geburtstag zu erleben, wenn ihr systolischer Blutdruck < 130 mmHg liegt. Das scheint selbst für 80-Jährige noch zu gelten.

Frauen bekommen seltener eine intensive Statintherapie

30.04.2024 Statine Nachrichten

Frauen in den Niederlanden erhalten bei vergleichbarem kardiovaskulärem Risiko seltener eine intensive Statintherapie als Männer. Ihre LDL-Zielwerte erreichen sie aber fast ähnlich oft.

Reizdarmsyndrom: Diäten wirksamer als Medikamente

29.04.2024 Reizdarmsyndrom Nachrichten

Bei Reizdarmsyndrom scheinen Diäten, wie etwa die FODMAP-arme oder die kohlenhydratreduzierte Ernährung, effektiver als eine medikamentöse Therapie zu sein. Das hat eine Studie aus Schweden ergeben, die die drei Therapieoptionen im direkten Vergleich analysierte.

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.