Waiting
Login-Verarbeitung ...

Trial ends in Request Full Access Tell Your Colleague About Jove

Cancer Research

Establishment and Characterization of Patient-Derived Xenograft Models of Anaplastic Thyroid Carcinoma and Head and Neck Squamous Cell Carcinoma

Published: June 2, 2023 doi: 10.3791/64623

Summary

The present protocol establishes and characterizes a patient-derived xenograft (PDX) model of anaplastic thyroid carcinoma (ATC) and head and neck squamous cell carcinoma (HNSCC), as PDX models are rapidly becoming the standard in the field of translational oncology.

Abstract

Patient-derived xenograft (PDX) models faithfully preserve the histological and genetic characteristics of the primary tumor and maintain its heterogeneity. Pharmacodynamic results based on PDX models are highly correlated with clinical practice. Anaplastic thyroid carcinoma (ATC) is the most malignant subtype of thyroid cancer, with strong invasiveness, poor prognosis, and limited treatment. Although the incidence rate of ATC accounts for only 2%-5% of thyroid cancer, its mortality rate is as high as 15%-50%. Head and neck squamous cell carcinoma (HNSCC) is one of the most common head and neck malignancies, with over 600,000 new cases worldwide each year. Herein, detailed protocols are presented to establish PDX models of ATC and HNSCC. In this work, the key factors influencing the success rate of model construction were analyzed, and the histopathological features were compared between the PDX model and the primary tumor. Furthermore, the clinical relevance of the model was validated by evaluating the in vivo therapeutic efficacy of representative clinically used drugs in the successfully constructed PDX models.

Introduction

The PDX model is an animal model in which human tumor tissue is transplanted into immunodeficient mice and grows in the environment provided by the mice1. Traditional tumor cell line models suffer from several disadvantages, such as the lack of heterogeneity, the inability to retain the tumor microenvironment, the vulnerability to genetic variations during repeated in vitro passages, and the poor clinical application2,3. The main drawbacks of genetically engineered animal models are the potential loss of the genomic features of human tumors, the introduction of new unknown mutations, and the difficulty in identifying the degree of homology between mouse tumors and human tumors4. In addition, the preparation of genetically engineered animal models is expensive, time-consuming, and relatively inefficient4.

The PDX model has many advantages over other tumor models in terms of reflecting tumor heterogeneity. From the perspective of histopathology, although the mouse counterpart replaces the human stroma over time, the PDX model preserves the morphological structure of the primary tumor well. In addition, the PDX model conserves the metabolomic identity of the primary tumor for at least four generations and better reflects the complex inter-relationships between tumor cells and their microenvironment, making it unique in simulating the growth, metastasis, angiogenesis, and immunosuppression of human tumor tissue5,6,7. At the cellular and molecular levels, the PDX model accurately reflects the inter- and intra-tumor heterogeneity of human tumors, as well as the phenotypic and molecular characteristics of original cancer, including gene expression patterns, mutation status, copy number, and DNA methylation and proteomics8,9. PDX models with different passages have the same sensitivity to drug therapy, indicating that the gene expression of PDX models is highly stable10,11. Studies have shown an excellent correlation between the response of the PDX model to a drug and the clinical responses of patients to that drug12,13. Therefore, the PDX model has emerged as a powerful preclinical and translational research model, particularly for drug screening and clinical prognosis prediction.

Thyroid cancer is a common malignant tumor of the endocrine system and is a human malignancy that has shown a rapid increase in incidence in recent years14. Anaplastic thyroid carcinoma (ATC) is the most malignant thyroid cancer, with a median patient survival of only 4.8 months15. Although only a minority of thyroid cancer patients are diagnosed with ATC each year in China, the mortality rate is close to 100%16,17,18. ATC usually grows rapidly and invades the adjacent tissues of the neck as well as the cervical lymph nodes, and about half of the patients have distant metastases19,20. Head and neck squamous cell carcinoma (HNSCC) is the sixth most common cancer in the world and one of the leading causes of cancer deaths, with an estimated 600,000 people suffering from HNSCC each year21,22,23. HNSCC includes a large number of tumors, including those in the nose, sinuses, mouth, tonsils, pharynx, and larynx24. ATC and HNSCC are two of the main head and neck malignancies. In order to facilitate the development of novel therapeutic agents and personalized treatments, it is necessary to develop robust and advanced preclinical animal models such as PDX models of ATC and HNSCC.

This article introduces detailed methods for establishing the subcutaneous PDX model of ATC and HNSCC, analyzes the key factors affecting the tumor take rate in model construction, and compares the histopathological characteristics between the PDX model and the primary tumor. Meanwhile, in this work, in vivo pharmacodynamic tests were performed using the successfully constructed PDX models in order to validate their clinical relevance.

Subscription Required. Please recommend JoVE to your librarian.

Protocol

All the animal experiments were performed in accordance with the Association for Assessment and Accreditation of Laboratory Animal Care guidelines and protocols approved by the Institutional Animal Care and Use Committee of West China Hospital, Sichuan University. NOD-SCID immunodeficient mice aged 4-6 weeks old (of both sexes) and female Balb/c nude mice aged 4-6 weeks old were used for the present study. The animals were obtained from a commercial source (see Table of Materials). The ethics committee of West China Hospital authorized the study with human subjects (protocol number 2020353). Each patient provided written informed consent.

1. Experimental preparation

  1. Arrange disposable blades, sterilized scissors and tweezers, and other instruments required for tumor transplantation, place them on the ultra-clean workbench, and irradiate them with ultraviolet light in advance.
  2. Prepare sterile saline and Petri dishes for use during the test.

2. Acquisition and transport of fresh tumor tissue

  1. Obtain fresh tumor samples (usually larger than 5 mm x 5 mm in size) from the operating room, and place them in a 15 mL or 50 mL centrifuge tube containing sterile HTK solution (see Table of Materials) or saline. Label the centrifuge tubes.
    NOTE: Fresh tumor samples were obtained by surgical removal or puncture from patients with ATC or HNSCC.
  2. Put the centrifuge tubes in an ice box prepared in advance.
    NOTE: During this time, the transplant operator must prepare the necessary items for transplantation (see Table of Materials).
  3. Ensure that the time between sample collection and transportation to the laboratory for PDX construction does not exceed 2 h. During transport, surround the tubes containing the tissues with an ice-water mixture or ice packs to preserve the tissue activity.

3. Tumor transplantation

  1. Once the tumor tissues arrive at the laboratory, record and renumber them.
    NOTE: For the present study, the patient information was kept strictly confidential. The remaining steps of the procedure were performed in a biosafety level 2 (BSL-2) laboratory. When entering the laboratory, wearing a smock over the work clothes or protective clothing, a hat, and a mask is recommended. The treatment of tumor tissue is conducted in a biosafety cabinet.
  2. Disinfect the centrifuge tubes containing the tumor tissues with 75% alcohol, and place them on the operating table. Transfer the tumor tissues to 6 cm Petri dishes filled with saline using sterilized ophthalmic forceps. Next, cut them into small pieces of about 2 mm x 2 mm and 3 mm x 3 mm using a blade.
  3. Transfer the pieces of tumor tissues into a 6 cm Petri dish containing the appropriate amount of saline, wrap the dish with the sealing film, place it in an ice box, and carry it into the specific pathogen-free (SPF) animal room along with the necessary instruments (a pair of scissors, forceps, and inoculation needles).
  4. Prepare the animal following the steps below.
    1. Remove the hair on the right lateral thorax of 4-6 week-old female or male NOD-SCID immunodeficient mice, and disinfect the skin with 75% alcohol. Anesthetize the mice by an intraperitoneal injection of 80 mg/kg ketamine and 10 mg/kg xylazine (see Table of Materials), and smear their eyes with vet ointment to prevent dryness. Confirm anesthesia depth via loss of pedal reflex.
    2. Make a 2 mm incision with scissors through the skin at the middle of the right lateral thorax of mice.
  5. Take a tumor piece from the Petri dish, and place it into the 2.4 mm x 2.0 mm trocar needle (see Table of Materials) with forceps.
  6. Hold the mouse, tighten the skin at the puncture site, use the trocar containing the tumor pieces to insert the tumor through the initial 2 mm skin incision, move to the back of the shoulder, and push the trocar core.
  7. Ensure that the tumor piece is pushed out and is left in the transitional sinus formed by the trocar puncture, and then pull out the trocar.
  8. If the tumor moves with the needle when it is withdrawn, use the trocar to reset it and suture the incision.
    NOTE: In this study, each mouse was inoculated at the dorsal fore and hind limbs. One to three mice were inoculated per tumor sample from each patient based on the tumor size.

4. Tumor tissue preservation, fixation, and protein freezing

NOTE: The remaining tumor tissues were used for seed preservation, fixation, and DNA/RNA/protein freezing, respectively.

  1. Remove the saline from the tumor surface with a sterile gauze before placing it in the cryopreservation tube to ensure that the tumor surface is not excessively wet.
  2. Put four to six pieces of 2 mm x 2 mm tumor tissue in a 2 mL cell cryopreservation tube, add 1 mL of cryopreservation solution composed of 90% fetal bovine serum (FBS) and 10% dimethyl sulfoxide (DMSO) into the tube, put the tube in a gradient cooling box, freeze it at −80 °C overnight, and finally, transfer it to liquid nitrogen.
  3. Place the 3 mm x 3 mm tumor tissue blocks in 10% buffered formalin for tissue fixation for pathological examination.
  4. Put the 3 mm x 3 mm tissue block into a 2 mL cell cryopreservation tube, quickly freeze it in liquid nitrogen, and then transfer to a −80 °C refrigerator for DNA/RNA and protein extraction.
  5. Collect the clinical information of the patients, such as the smoking history, tumor size, differentiation, pathological subtype, cancer grade, cancer stage, distant metastasis, origin, medical history, immunohistochemistry, human papillomavirus (HPV) infection in HNSCC patients, and treatment medication.

5. Passaging, cryopreservation, and resuscitation of PDX model tumors

  1. Measure the length and width of the subcutaneous tumors in mice by using vernier calipers once a week, and calculate the tumor volume according to the formula: tumor volume = 0.5 × length × width2. Draw the tumor growth curve.
  2. When the PDX tumor reaches 2,000 mm3, passage it to the next generation of mice, and perform tumor re-transplantation. Perform the preparation of the instruments following step 4.
  3. Euthanize the mice by cervical dislocation after anesthetizing with 80 mg/kg ketamine.
  4. Disinfect the skin with 75% alcohol. Then, cut the skin surrounding the tumor using scissors, then remove the tumor with forceps, and place it in a Petri dish. 
  5. Perform the tumor transplantation procedure following step 3.
  6. Perform the preservation and cryopreservation of the PDX model tumors following step 4.
  7. For the resuscitation of the tumor tissue, follow the principle of slow freezing and quick dissolving. After taking out the cryovials from liquid nitrogen, quickly place them in a water bath at 37 °C for rapid dissolving.
  8. Gently shake the cryovials in the water bath to accelerate the thawing process.
  9. Thaw, transfer the tumor pieces to the prepared normal saline for washing, and then inoculate the next generation of mice. For the specific operation, please see the tissue transplantation procedure in step 3.

6. Determining the therapeutic efficacy of lenvatinib and cisplatin in the ATC PDX model

NOTE: The ATC PDX model was used to test the therapeutic effect of the tyrosine kinase inhibitor lenvatinib and the chemotherapeutic drug cisplatin25,26,27.

  1. Select the P5 generation tumor tissue of an ATC PDX model (THY-017), cut into 2-4 mm3 tissue pieces, and inoculate subcutaneously (step 3) to the right back of ten 4-6 week female Balb/c nude mice.
  2. Select 15 mice with tumor volumes between 50-150 mm3, and divide them into three groups.
  3. Administer lenvatinib (10 mg/kg) intragastrically to one group once daily for 15 days, administer cisplatin (3 mg/kg) intraperitoneally to one group every 3 days for a total of six doses, and administer the control group with the same volume of normal saline.
  4. Measure the body weight and tumor volume of the mice twice per week.
  5. At the end of the test, euthanize the mice (step 5.3), and weigh the tumors.

Subscription Required. Please recommend JoVE to your librarian.

Representative Results

A total of 18 thyroid cancer specimens were transplanted, and five PDX models of thyroid cancer were successfully constructed (27.8% tumor take rate), including four cases of undifferentiated thyroid cancers and one case of anaplastic thyroid cancer. The correlation between the success rate of model construction and the age, gender, tumor diameter, tumor grade, and differentiation were analyzed. Although the model success rate of grade 4 tumor samples was higher than for samples with lower grades, and the success rate of undifferentiated tumor samples was also higher than that of highly differentiated samples, the correlation analysis results showed that these factors were not associated with the success rate of the PDX model (Table 1). Seventeen HNSCC samples were inoculated, and four PDX models of HNSCCC were successfully constructed. The correlation analysis between the tumor take rate in the model construction and the clinical parameters of the tumor samples demonstrated that the degree of differentiation was associated with model success rate, while age, gender, smoking history, tumor diameter, cancer grade, metastasis, and HPV infection did not affect the tumor take rate (Table 2).

The tumor growth curves for each PDX model were plotted to better understand the growth rates of the PDX models from different patients (Figure 1, Figure 2, and Table 3). The average tumorigenic cycles (time from inoculation to a tumor size of 1,000 mm3) of THY-004 from generations P0 to P5 were 68 days, 87 days, 29 days, 34 days, 28 days, and 26 days, respectively. The average tumorigenic cycles of THY-012 from generations P0 to P5 were 119 days, 61 days, 66 days, 55 days, 87 days, and 116 days, respectively. The average tumorigenic cycles of THY-017 from generations P0 to P5 were 27 days, 17 days, 30 days, 13 days, 22 days, and 15 days, respectively. The average tumorigenic cycles of THY-018 from generations P0 to P3 were 134 days, 70 days, 48 days, and 48 days, respectively. The average tumorigenic cycles of THY-021 from generations P0 to P3 were 53 days, 66 days, 35 days, and 49 days, respectively. The average tumorigenic cycles of OTO-017 from generations P0 to P4 were 118 days, 86 days, 67 days, 129 days, and 88 days, respectively. The average tumorigenic cycles of OTO-022 from generations P0 to P5 were 155 days, 55 days, 32 days, 37 days, 27 days, and 46 days, respectively. The average tumorigenic cycles of OTO-030 from generations P0 to P2 were 133 days, 93 days, and 104 days, respectively. The average tumorigenic cycles of OTO-031 from generations P0 to P5 were 144 days, 58 days, 33 days, 34 days, 52 days, and 50 days, respectively. The ATC samples were stably passed to the P3 generation and later, while two cases of HNSCC samples failed to form tumors after passing to the P1 generation. The growth rates of some samples were relatively slow in the P0 generation, but their growth rates were accelerated after passing to the P1 and later generations. The histopathological characteristics of the patient tumors with those of different generations of PDX models were compared. The results showed that PDX tumors and patient-derived primary tumors were morphologically almost similar (Figure 3), with slight differences that may be due to the heterogeneity in the sampling area between patients and different generations of PDX.

The anti-tumor efficacy of lenvatinib (a multi-target tyrosine kinase inhibitor approved for the treatment of advanced thyroid cancer28) was evaluated in the PDX model of ATC. As shown in Figure 4A, lenvatinib treatment significantly inhibited the tumor growth in the ATC PDX model compared to the normal saline control group (P < 0.05). At the end of the experiment, the tumor tissue was excised and weighed to determine the tumor weight. Compared with the control group, the tumor weight of the lenvatinib treatment group was lower, although a statistical difference was not achieved (Figure 4B). In addition, no obvious changes in general status and body weight were observed in the mice treated with lenvatinib (Figure 4C). Due to the excessive frequency of cisplatin administration during the experiments, the mice showed significant toxicity, manifested by weight loss and even death. The anti-tumor efficacy of cisplatin is shown in Supplementary Figure 1.

Figure 1
Figure 1: Tumor growth curve of ATC PDX models from different patients. Each color represents the specified generation, and each curve represents a single tumor. One to three mice were inoculated at the passage 0 (P0) generation, and five mice were inoculated in the subsequent passages (P1-P5). Please click here to view a larger version of this figure.

Figure 2
Figure 2: Tumor growth curve of the HNSCC PDX models from different patients. Each color represents the specified generation, and each curve represents a single tumor. One to three mice were inoculated at the P0 generation, and five mice were inoculated at P1 and later generations. Please click here to view a larger version of this figure.

Figure 3
Figure 3: Histopathological study. Histopathological comparison between the patient primary tumors and corresponding PDXs (passage 1 and passage 3) of ATC (THY-012, THY-017) and HNSCC (OTO-017) (hematoxylin-eosin staining, 100x). The pathological subtypes of THY-012 and THY-017 were anaplastic thyroid carcinoma, and the pathological subtype of OTO-017 was squamous cell carcinoma. Scale bars = 100 µm. Please click here to view a larger version of this figure.

Figure 4
Figure 4: The therapeutic efficacy of lenvatinib in the ATC PDX model. Changes in the (A) tumor volume, (B) tumor weight, and (C) body weight of ATC PDX-bearing mice after treatment with lenvatinib (10 mg/kg). Statistical analyses were performed using T-tests to compare levatinib with control. *P < 0.05 versus control was considered to be statistically significant. Please click here to view a larger version of this figure.

Parameters Class Tumor take rate (%) P
Age (years) <60 16.67 (1/6) 0.615
≥60 33.33 (4/12)
Gender Male 16.67 (1/6) 0.615
Female 33.33 (4/12)
Tumor diameter <6cm 37.50 (3/8) 0.608
≥6cm 20.00 (2/10)
Pathologic TNM stage I 0.00 (0/1) 1
Ш 0.00 (0/1)
Equation 2 31.25 (5/16)
Differentiation High 0.00 (0/7) 0.059
Poor 25.00 (1/4)
Undifferentiated 57.14 (4/7)

Table 1: Correlation between the ATC tumor take rate and the clinical characteristics of the patients.

Parameters Class Tumor take rate (%) P
HPV Negative 33.33 (2/6) 1
Unknown or positive 36.36 (4/11)
Age (years) <60 33.33 (3/9) 1
≥60 37.50 (3/8)
Gender Male 50.00 (5/10) 0.304
Female 14.29 (1/7)
Smoking status Ever 44.44 (4/9) 0.62
Never 25.00 (2/8)
Tumor diameter <3cm 40.00 (4/10) 1
≥3cm 28.57 (2/7)
Pathologic TNM stage I 75.00 (3/4) 0.423
Equation 1 25.00 (2/8)
Ш 0.00 (0/1)
Equation 2 33.33 (1/3)
Distant metastasis Y 28.57 (2/7) 0.633
N 44.44 (4/9)
Differentiation High 12.50 (1/8) 0.036*
Moderate to high 100.00 (2/2)
Moderate 0.00 (0/2)
Moderate to poor 66.67 (2/3)
* P < 0.05

Table 2: Correlation between the HNSCC tumor take rate and the clinical characteristics of the patients. *P < 0.05.

Sample name Generation P to P0 Generation P0 to P1 Generation P1 to P2 Generation P2 to P3 Generation P3 to P4 Generation P4 to P5
THY-004 68 87 29 34 28 26
THY-012 119 61 66 55 87 116
THY-017 27 17 30 13 22 15
THY-018 134 70 48 48 - -
THY-021 53 66 35 49 - -
OTO-017 118 86 67 129 - -
OTO-022 155 55 32 37 27 46
OTO-030 133 93 104 - - -
OTO-031 144 58 33 34 52 50

Table 3: The average tumorigenic cycle (time from inoculation to a tumor size of 1,000 mm3) of the ATC and HNSCC models.

Supplementary Figure 1: The therapeutic efficacy of cisplatin in the ATC PDX model. Changes in the (A) tumor volume, (B) tumor weight, and (C) body weight of ATC PDX-bearing mice after treatment with cisplatin (3 mg/kg). Statistical analyses were performed using T-tests to compare cisplatin with the control. *P < 0.05 versus control was considered to be statistically significant. Please click here to download this File.

Subscription Required. Please recommend JoVE to your librarian.

Discussion

This study has successfully established the subcutaneous PDX models of ATC and HNSCC. There are many aspects to pay attention to during the process of PDX model construction. When the tumor tissue is separated from the patient, it should be put into the ice box and sent to the laboratory for inoculation as soon as possible. After the tumor arrives at the laboratory, the operator must pay attention to maintaining a sterile field and practice aseptic procedures. For needle biopsy samples, because the tumor tissue is particularly small, inoculation after mixing the sample with the matrix gel would be more conducive to establishing the model. The primary tumor tissue should also be preserved, fixed, and frozen as much as possible for future research. During inoculation, the air in the trocar needs to be expelled as much as possible after the tumor pieces have been put into the trocar before use. After tumor inoculation, the tumor growth should be observed in mice for 1-4 months, and mice without tumor growth for more than 6 months can be euthanized29.

Immunodeficient mice are generally chosen as the host for PDX model construction29,30. From the P0 generation to the P2 generation, non-obese diabetic-severely compromised immune deficient (NOD-SCID) mice or NOD Cg-Prkdcscid Il2rgtm1Wjl/SzJ (NSG) mice are generally used. In the P3 generation and beyond, the samples are considered to be stably passed, so nude mice can usually also serve as the host, and tumors can also grow normally. Additionally, the total operation time, tumor isolation time, disease-free survival, and overall survival rate of the patients, the tumor malignant degree, and the histologic subtype were all associated with PDX model tumorigenicity31,32,33,34. The transplantation site also has an impact on the success rate of PDX modeling, and studies have shown that renal capsule and orthotropic transplantation have a high tumorigenic rate33,35. In addition, the use of Matrigel may also improve the tumorigenic rate36,37. It has been reported that human papillomavirus (HPV) infection affects the success rate of transplantation in HNSCC tumors; HPV-negative tumors have a superior take rate compared to HPV-positive tumors38,39. This study did not reach the same conclusion, probably due to the small sample numbers and incomplete information on HPV infection.

Different from the orthotopic and renal capsule transplantation models, the subcutaneous model is more convenient for observing the growth of tumors and is also conducive to operation40,41,42. Based on the tumor growth data of the ATC and HNSCC PDX model, we found that the growth rates of tumors from different patients were inconsistent, reflecting inter-tumor heterogeneity. The tumor growth rate of the P0 generation from most PDX models was relatively slower than for the latter passages, which was likely due to the adaption of the mouse microenvironment. Notably, the growth rate from some patient-derived tumors increased in different passages after the P1 generation, consistent with the shortened passage interval reported by Pearson et al.43. Histopathological examination demonstrated that the PDX tumors retained the morphological characteristics of the primary tumors. The correlation between the PDX model and the clinical ATC patients was also reflected in the results of the in vivo pharmacodynamic tests, which demonstrated that Lenvartinib exhibited a good anti-tumor effect, consistent with clinical reports25,26,27.

However, the PDX model also has certain disadvantages. For example, the tumor formation time is relatively long, which is unsuitable for patients with advanced or aggressive tumors. In addition, the time and monetary costs of high-throughput drug screening are too high44. Indeed, combining the PDX model with tumor organoids and establishing a patient-derived organoid (PDO) model corresponding to the PDX model would compensate for this deficiency44,45,46. Orthotopic transplantation models can be used to study the pathogenesis and metastatic mechanisms of tumors40,41,47. The lack of a functional immune system is another disadvantage of the PDX model, so increasing numbers of experiments are using humanized mice to construct the PDX model for tumor immunology research48,49,50.

Subscription Required. Please recommend JoVE to your librarian.

Disclosures

No potential conflicts of interest are disclosed.

Acknowledgments

This work was supported by the Sichuan Province Science and Technology Support Program (Grant Nos. 2019JDRC0019 and 2021ZYD0097), the 1.3.5 project for disciplines of excellence, West China Hospital, Sichuan University (Grant No. ZYJC18026), the 1.3.5 project for disciplines of excellence-Clinical Research Incubation Project, West China Hospital, Sichuan University (Grant No. 2020HXFH023), the Fundamental Research Funds for the Central Universities (SCU2022D025), the International Cooperation Project of Chengdu Science and Technology Bureau (Grant No. 2022-GH02-00023-HZ), the Innovation Spark Project of Sichuan University (Grant No. 2019SCUH0015), and the Talent Training Fund for Medical-engineering Integration of West China Hospital - University of Electronic Science and Technology (Grant No. HXDZ22012).

Materials

Name Company Catalog Number Comments
2.4 mm x 2.0 mm trocar Shenzhen Huayang Biotechnology Co., Ltd 18-9065
Balb/c nude mice Beijing Vital River Laboratory Animal Technology Co., Ltd. 401
Biosafety cabinet Suzhou Antai BSC-1300IIA2
Blade Shenzhen Huayang Biotechnology Co., Ltd 18-0823
Centrifuge tube  Corning 430791/430829
Cryopreservation tube Chengdu Dianrui Experimental Instrument Co., Ltd /
Custodiol HTK-Solution Custodiol 2103417
Dimethyl sulfoxide(DMSO) SIGMA-ALORICH D5879-500mL
Electronic balance METTLER ME104
Electronic digital caliper Chengdu Chengliang Tool Group Co., Ltd 0-220
fetal bovine serum(FBS) VivaCell C04001-500
IBM SPSS Statistics 26 IBM
Ketamine Jiangsu Zhongmu Beikang Pharmaceutical Co., Ltd  100761663
Lenvatinib ApexBio A2174
NOD-SCID immunodeficient mice Beijing Vital River Laboratory Animal Technology Co., Ltd. 406
Pen-Strep Solution Biological Industries 03-03101BCS
Petri dish WHB WHB-60/WHB-100
Saline  Sichuan Kelun W220051705
Scissor Shenzhen Huayang Biotechnology Co., Ltd 18-0110
Tweezer Shenzhen Huayang Biotechnology Co., Ltd 18-1241
Vet ointment Pfizer Inc. P10015353
Xylazine Dunhua Shengda Animal Medicine Co., Ltd 070031777

DOWNLOAD MATERIALS LIST

References

  1. Toolan, H. W. Successful subcutaneous growth and transplantation of human tumors in X-irradiated laboratory animals. Proceedings of The Society for Experimental Biology and Medicine. 77 (3), 572-578 (1951).
  2. Gillet, J. P., et al. Redefining the relevance of established cancer cell lines to the study of mechanisms of clinical anti-cancer drug resistance. Proceedings of the National Academy of Sciences of the United States of America. 108 (46), 18708-18713 (2011).
  3. Hausser, H. J., Brenner, R. E. Phenotypic instability of Saos-2 cells in long-term culture. Biochemical & Biophysical Research Communications. 333 (1), 216-222 (2005).
  4. Pérez-Mancera, P., Guerra, C., Barbacid, M., Tuvesonet, D. A. What we have learned about pancreatic cancer from mouse models. Gastroenterology. 142 (5), 1079-1092 (2012).
  5. Bruna, A., et al. A biobank of breast cancer explants with preserved intra-tumor heterogeneity to screen anticancer compounds. Cell. 167 (1), 260-274 (2016).
  6. Choi, S., et al. Lessons from patient-derived xenografts for better in vitro modeling of human cancer. Advanced Drug Delivery Reviews. 79-80, 222-237 (2014).
  7. Blomme, A., et al. Murine stroma adopts a human-like metabolic phenotype in the PDX model of colorectal cancer and liver metastases. Oncogene. 37 (9), 1237-1250 (2018).
  8. Wang, D., et al. Molecular heterogeneity of non-small cell lung carcinoma patient-derived xenografts closely reflect their primary tumors. International Journal of Cancer. 140 (3), 662-673 (2016).
  9. Jung, J., et al. Generation and molecular characterization of pancreatic cancer patient-derived xenografts reveals their heterologous nature. Oncotarget. 7 (38), 62533-62546 (2016).
  10. Keysar, S., et al. A patient tumor transplant model of squamous cell cancer identifies PI3K inhibitors as candidate therapeutics in defined molecular bins. Molecular Oncology. 7 (4), 776-790 (2013).
  11. Rubio-Viqueira, B., et al. An in vivo platform for translational drug development in pancreatic cancer. Clinical Cancer Research. 12 (15), 4652-4661 (2006).
  12. Fiebig, H. H., et al. Development of three human small cell lung cancer models in nude mice. Recent Results in Cancer Research. 97, 77-86 (1985).
  13. Morelli, M. P., et al. Prioritizing phase I treatment options through preclinical testing on personalized tumorgraft. Journal of Clinical Oncology. 30 (4), 45-48 (2012).
  14. Bray, F., et al. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA. 68 (6), 394-424 (2018).
  15. Onoda, N., et al. Evaluation of the 8th edition TNM classification for anaplastic thyroid carcinoma. Cancers. 12 (3), 552 (2020).
  16. Nel, C., et al. Anaplastic carcinoma of the thyroid: A clinicopathologic study of 82 cases. Mayo Clinic Proceedings. 60 (1), 51-58 (1985).
  17. Mazzaferri, E. L. Increasing incidence of thyroid cancer in the United States, 1973-2002. Yearbook of Medicine. 2007, 496-499 (2007).
  18. Kebebew, E., Greenspan, F. S., Clark, O. H., Woeber, K. A., Mcmillan, A. Anaplastic thyroid carcinoma. Treatment outcome and prognostic factors. Cancer. 103 (7), 1330-1335 (2005).
  19. Lin, B., et al. The incidence and survival analysis for anaplastic thyroid cancer: A SEER database analysis. American Journal of Translational Research. 11 (9), 5888-5896 (2019).
  20. Maniakas, A., Dadu, R., Busaidy, N. L., Wang, J. R., Zafereo, M. Evaluation of overall survival in patients with anaplastic thyroid carcinoma, 2000-2019. JAMA Oncology. 6 (9), 1397-1404 (2020).
  21. Gilardi, M., et al. Tipifarnib as a precision therapy for HRAS-mutant head and neck squamous cell carcinomas. Molecular Cancer Therapeutics. 19 (9), 1784-1796 (2020).
  22. Siegel, R. L., Miller, K. D., Jemal, A. Cancer statistics, 2016. CA. 66 (1), 7-30 (2016).
  23. Chow, L. Q. M. Head and neck cancer. New England Journal of Medicine. 382 (1), 60-72 (2020).
  24. Swiecicki, P. L., Brennan, J. R., Mierzwa, M., Spector, M. E., Brenner, J. C. Head and neck squamous cell carcinoma detection and surveillance: Advances of liquid biomarkers. Laryngoscope. 129 (8), 1836-1843 (2019).
  25. Wang, R., et al. Distribution and activity of lenvatinib in brain tumor models of human anaplastic thyroid cancer cells in severe combined immune deficient mice. Molecular Cancer Therapeutics. 18 (5), 947-956 (2019).
  26. Takahashi, S., et al. A phase II study of the safety and efficacy of lenvatinib in patients with advanced thyroid cancer. Future Oncology. 15 (7), 717-726 (2019).
  27. Ferrari, S. M., et al. Lenvatinib exhibits antineoplastic activity in anaplastic thyroid cancer in vitro and in vivo. Oncology Reports. 39 (5), 2225-2234 (2018).
  28. Cabanillas, M. E., Habra, M. A. Lenvatinib: Role in thyroid cancer and other solid tumors. Cancer Treatment Reviews. 42, 47-55 (2016).
  29. Jung, J., Seol, H. S., Chang, S. The generation and application of patient-derived xenograft model for cancer research. Cancer Research and Treatment. 50 (1), 1-10 (2018).
  30. Peng, S., et al. Tumor grafts derived from patients with head and neck squamous carcinoma authentically maintain the molecular and histologic characteristics of human cancers. Journal of Translational Medicine. 11, 198 (2013).
  31. Derose, Y. S., et al. Tumor grafts derived from women with breast cancer authentically reflect tumor pathology, growth, metastasis and disease outcomes. Nature Medicine. 17 (11), 1514-1520 (2011).
  32. Chen, X., Shen, C., Wei, Z., Zhang, R., Xiao, K. Patient-derived non-small cell lung cancer xenograft mirrors complex tumor heterogeneity. Cancer Biology and Medicine. 18 (1), 184-198 (2021).
  33. Choi, Y. Y., et al. Establishment and characterisation of patient-derived xenografts as paraclinical models for gastric cancer. Scientific Reports. 6, 22172 (2016).
  34. Maider, I. V., Andrés, C., Alberto, B. Preclinical models for precision oncology. Biochimica et Biophysica Acta (BBA) - Reviews on Cancer. 1872 (2), 239-246 (2018).
  35. Okada, S., Vaeteewoottacharn, K., Kariya, R. Establishment of a patient-derived tumor xenograft model and application for precision cancer medicine. Chemical & Pharmaceutical Bulletin. 66 (3), 225-230 (2018).
  36. Michael, G., et al. Tumor take rate optimization for colorectal carcinoma patient-derived xenograft models. BioMed Research International. 2016, 1715053 (2016).
  37. Bernardo, C., Costa, C., Sousa, N., Amado, F., Santos, L. Patient-derived bladder cancer xenografts: a systematic review. Translational Research. 166 (4), 324-331 (2015).
  38. Facompre, N. D., et al. Barriers to generating PDX models of HPV-related head and neck. Laryngoscope. 127 (12), 2777-2783 (2017).
  39. Kang, H. N., Kim, J. H., Park, A. Y., Choi, J. W., Kim, H. R. Establishment and characterization of patient-derived xenografts as paraclinical models for head and neck cancer. BMC Cancer. 20 (1), 316 (2020).
  40. Ahn, S. H., et al. An orthotopic model of papillary thyroid carcinoma in athymic nude mice. Archives of Otolaryngology-Head & Neck Surgery. 134 (2), 190-197 (2008).
  41. Nucera, C., et al. A novel orthotopic mouse model of human anaplastic thyroid carcinoma. Thyroid. 19 (10), 1077-1084 (2009).
  42. De Rose, F., et al. Galectin-3 targeting in thyroid orthotopic tumors opens new ways to characterize thyroid cancer. Journal of Nuclear Medicine. 60 (6), 770-776 (2019).
  43. Pearson, A. T., et al. Patient-derived xenograft (PDX) tumors increase growth rate with time. Oncotarget. 7 (7), 7993-8005 (2016).
  44. Huo, K. G., D'Arcangelo, E., Tsao, M. S. Patient-derived cell line, xenograft and organoid models in lung cancer therapy. Translational Lung Cancer Research. 9 (5), 2214-2232 (2020).
  45. Kumari, R., Xu, X., Li, H. Q. Translational and clinical relevance of PDX-derived organoid models in oncology drug discovery and development. Current Protocols. 2 (7), e431 (2022).
  46. Takahashi, N., et al. Construction of in vitro patient-derived tumor models to evaluate anticancer agents and cancer immunotherapy. Oncology Letters. 21 (5), 406 (2021).
  47. Barasch, A., et al. Photobiomodulation effects on head and neck squamous cell carcinoma (HNSCC) in an orthotopic animal model. Supportive Care in Cancer. 28 (6), 2721-2727 (2020).
  48. Wang, M., et al. Humanized mice in studying efficacy and mechanisms of PD-1-targeted cancer immunotherapy. FASEB Journal. 32 (3), 1537-1549 (2018).
  49. Wu, C., Wang, X., Shang, H., Wei, H. Construction of a humanized PBMC-PDX model to study the efficacy of a bacterial marker in lung cancer immunotherapy. Disease Markers. 2022, 1479246 (2022).
  50. Yao, L. C., et al. Creation of PDX-bearing humanized mice to study immuno-oncology. Methods in Molecular Biology. 1953, 241-252 (2019).

Tags

Patient-derived Xenograft Models Anaplastic Thyroid Carcinoma Head And Neck Squamous Cell Carcinoma Tumor Heterogeneity Molecular Diversity Clinic Outcomes Anti-cancer Drug Sensitivity Screening Personalized Treatment ATC And HNSCC Tumor Samples HTK Solution Disinfection Ophthalmic Forceps Petri Dish Saline Cutting Tumor Tissue Sealing Film Pathogen-free Animal Room Inoculation Needles Anesthetizing The Mouse Right Lateral Thorax
Establishment and Characterization of Patient-Derived Xenograft Models of Anaplastic Thyroid Carcinoma and Head and Neck Squamous Cell Carcinoma
Play Video
PDF DOI DOWNLOAD MATERIALS LIST

Cite this Article

Wu, M., Liu, Y., Zhao, Y., Zhang,More

Wu, M., Liu, Y., Zhao, Y., Zhang, Y., Huang, L., Du, Q., Zhang, T., Zhong, Z., Luo, H., Xiao, K. Establishment and Characterization of Patient-Derived Xenograft Models of Anaplastic Thyroid Carcinoma and Head and Neck Squamous Cell Carcinoma. J. Vis. Exp. (196), e64623, doi:10.3791/64623 (2023).

Less
Copy Citation Download Citation Reprints and Permissions
View Video

Get cutting-edge science videos from JoVE sent straight to your inbox every month.

Waiting X
Simple Hit Counter