Author: Dr. Yu Xiaofeng, senior vice president of tournament business model animal, Senior Scientist cutting edge The genetically modified mouse (GEM) model is a de novo tumor established under natural intact immune conditions relative to the tumor cell vaccination and immunodeficient mouse models commonly used in the past. Therefore, as a tool for oncology research, the GEM model can mimic the histopathological and molecular characteristics of human tumors, showing better genetic heterogeneity, which has the advantage of reflecting the tumor cells themselves and the tumor microenvironment. Interacting factors such as cells, including the ability to cause the primary tumor to begin to develop into a metastatic disease. The establishment and application of the GEM model has greatly promoted the development and progress of oncology research. At present, the GEM model has been successfully applied to verify potential tumor genes and drug targets, assess treatment effects, analyze the effects of tumor microenvironment, and evaluate drug resistance mechanisms. Moreover, combining clinical patient research and constructing a more reasonable and effective GEM model to optimize preclinical trials of tumor intervention will undoubtedly further promote the development of new strategies for tumor therapy and improve the success rate of its effective conversion into clinical application. The main challenges facing current oncology research Oncology research and tumor treatment still face many challenges in clinical practice. Among them, the formation of anti-tumor drug tolerance and tumor metastasis diseases are two important practical problems currently facing. Anti-tumor resistance is caused by the emergence of primary mutations in heterogeneous tumors, or by the massive growth of pre-treatment resistant clones, while existing monotherapy or chemical drug therapies targeting anti-tumor agents are available. Drug tolerance cannot be avoided. Moreover, after a clearly successful treatment, a small amount of drug-tolerant tumor cells can survive and become a major cell population after a certain period of time, eventually forming a recurrent disease that is different from the original tumor phenotype. Tumor metastatic disease is the cause of more than 90% of cancer-related deaths, because there are currently no effective treatments for these secondary tumors. In recent years, although some encouraging progress has been made in tumor immunotherapy for intervention in the immune system of cancer patients, the therapy has only been effective in some specific cases, and it has no practical clinical significance for most tumor patients. The so-called successful cancer treatment often requires synergistic effects of various methods, such as surgery, radiation exposure, cytotoxic therapy, and comprehensive strategies such as immunotherapy. In order to design effective and reasonable comprehensive treatment measures and programs, the primary basis and premise is to understand the formation and development of tumors, metastasis, and the mechanism of interaction between tumor cells themselves and their microenvironment cells in response to treatment, so as to find The most effective treatment for different tumor types. To achieve this, researchers must rely on preclinical studies of animal models. Although in the past it relied on traditional preclinical mouse models (ie, through the establishment of transplanted human tumor cell lines or homologous mouse tumor cell line models) to obtain successful validation of new preclinical anticancer therapies, the vast majority of these new The therapy ended in failure in the clinical phase III trial. In general, the traditional in vivo tumor mouse model does not perform well in predicting the effectiveness of new clinical therapies, thus highlighting the significance and value of finding improved preclinical models with better predictive power and effects. Recent advances and developments in genetic modification technology have made it possible to rapidly develop a GEM model that can more effectively simulate human tumors. The GEM model has a genetic composition, interaction between tumor cells and its tumor microenvironment, drug response and tolerance. Closer to the tumor patient. The emergence of a new generation of GEM models has also greatly promoted the transformation of new anti-tumor strategies into clinical applications, ultimately achieving the goal of improving the survival rate of cancer patients. Advantages and disadvantages of traditional mouse models commonly used in oncology research The mouse tumor transplantation model, which was first established in nude mice 50 years ago to transplant human/mouse tumor cell lines, has become a common mouse model for tumor research. This type of transplantation model can quickly test potential tumor and metastasis-related genes and become clinical. The main tool for pre-drug testing. For example, xenograft studies help to reveal the mechanism of colon cancer (CRC) tolerance to drugs (such as Vemurafenib) in mice, enabling the initiation of CRC patients in clinical trials against both mutant BRAF (eg V600E) and EGFR. The targeted combination therapy demonstrates the practical significance of this type of xenograft model in establishing a new combination therapy strategy. Xenograft studies are also helpful in identifying specific gene expression profiles and studying the characteristics of organ-specific localization and metastasis. The application of this model confirms that the spread of breast cancer cells is present in the vicinity of blood vessels, which provides a possible countermeasure for regulating these spread breast cancer cells. Moreover, in vivo model studies using tumor cell line transplantation have provided many basic concepts for anti-tumor immunity, T cell tolerance mechanisms, and tumor immune escape pathways. These findings have laid the foundation for the ongoing breakthrough in tumor immunotherapy. However, since tumor cell lines contain many mutations at the beginning and additional mutations are generated during long-term in vitro culture, such vaccination models are difficult to truly reflect the morphological and genetic heterogeneity of human tumor cells, thereby The reliability of this type of model as a predictive effect of clinical application is reduced. In addition, in order to prevent possible rejection, the xenograft model of tumor cell lines is based on immunodeficient mice, which also limits its application in the fields of immune system and therapeutic response in tumor development. Unlike cell line transplantation models, patient-derived tumor xenografts (PDTX, or PDX) models constructed by transplanting fresh human tumor biopsies into immunodeficient mice have more advantages, PDX mouse models. The tumor retains features of tumor tissue molecules, genetics, and histological heterogeneity from tumor patients (even after passage through several generations in mice). Therefore, the PDX model has also become a useful tool for personalized medicine and preclinical drug screening. At present, the research of PDX model has been applied to potential clinical drug prediction experiments on a large scale. Some researchers have constructed about 1000 PDX models from different types of mutations and applied these different PDX models to screen different drugs in mice to find the correlation between drugs and tumor genotypes to achieve experimental repeatability and Unification of clinical interpretability. Unfortunately, the dissatisfaction of the PDX model in studying certain types of tumors, such as estrogen receptor-positive breast cancer and prostate cancer, has become a major obstacle to its application. Moreover, the PDX model must be built on immunodeficient mice that lack the natural anti-tumor and tumor-promoting activities mediated by the acquired immune system. The researchers also understand that although the PDX model lacks a functional acquired immune system, the model can provide valuable clinical data. A corresponding improvement in progress is to reconstruct a humanized mouse model of the human immune system by transplanting human CD34-positive hematopoietic stem cells or precursor cells with significant success. Although the reconstruction of human immune cells from certain specific lineages in mice is still challenging, it can promote the development and maturation of human bone marrow cells in mice by introducing human-related cytokines, chemokines, and growth factors. Effect. To construct immunodeficient mice optimized to support human HLA-restricted T cell development, a humanized mouse model was established using genetic modification techniques to introduce human HLA molecules into the corresponding regions of knockout mouse MHC types I and II. The humanized mouse model can be used as a useful tool for preclinical evaluation of immunotherapy. However, when the source of the human hematopoietic cell donor (such as by cord blood or fetal liver) is limited, the higher construction cost in the actual operation also naturally becomes The disadvantages of the practical application of the model. Application of continuous improvement of GEM model to study primary tumor After the successful establishment of mouse pronuclear injection transgenic technology in the 1970s, in the early 1980s, the method was used to introduce the cloned oncogene into the mouse genome for the first time, and the so-called oncomice was successfully prepared. The oncogenic mouse is the first tumor GEM model that specifically expresses the oncogene v-HRas using a mammary gland specific promoter (MMTV), and the oncogenic mouse was successfully constructed for the first time, confirming that the mouse forms a primary breast tumor, and This has greatly inspired the field of cancer research, because the results of this study for the first time truly prove the hypothesis that oncogenes can express tumors in normal cells. In 1992, along with the breakthrough development of mouse embryonic stem cell (ES) gene targeting technology, the successful construction of tumor suppressor gene (TSG) knockout mice also proved the important role of this kind of TSG gene in tumorigenesis. Although carcinogenic mice and TSG knockout mice provide a very valuable theoretical basis, these two models also have their limitations. Since the mouse obtained by transgenic technology is to express the transferred foreign gene to all cells in a specific tissue, the TSG knockout mouse is a related gene in all cells in the body. However, the formation process of a real tumor is a diffuse tumor phenomenon caused by the accumulation of a certain genetic variation in a single cell under the premise of the health of the entire tissue and organ in the individual. In order to meet the actual conditions of the tumor formation process, it is more necessary to design and construct a more reasonable or complex mouse model, such as the conditional inactivation of tumor suppressor genes in somatic cells, or the activation (mutation) of oncogenes. GEM model. The basic principle of the construction of the conditional GEM model is to add the loxP recombination site to the two ends of the modified gene. In the presence of a specific Cre recombinase, the DNA between the ends of the loxP can be knocked out under certain conditions. The purpose of inactivating the gene. The first successful example of the application of this model is a mouse colorectal cancer model constructed using the Cre-loxP system-mediated somatic cell inactivation of the Apc gene. Adenoviral vector is used to express Cre recombinase specifically in intestinal epithelial cells, and tissue-specific knockout of APC gene causes rapid formation of scattered colorectal adenoma in mice, which is characterized by familial colon adenomatous polyposis (FAP) patients. There are many similarities. Therefore, by discovering mutants of specific cancer genes, researchers can construct mouse models that are more capable of mimicking the similar characteristics of tumor patients in histology, molecular science, and clinically. With the Cre-ERT fusion protein induction system, researchers can modify the relevant target genes in somatic cells at specific times with specific tissues, merging the mutant hormone binding region of the estrogen receptor with Cre recombinase to establish an inducible The regulated Cre recombinase expression system, when in the presence of an estrogen analog (such as Tamoxifen), causes Cre recombinase activity to be activated after translation, exerting its role in recognizing the loxP site, and achieving the purpose of inducible cleavage of the target gene. . Therefore, the conditional genetic modification of LoxP mice (ie, loxP sites on both ends of the target gene DNA), by adding the inducer Tamoxifen at the selected time, controls the specific expression of Cre-ERT to achieve the target gene. Time and space and the purpose of specific modifications on the area. Although the Cre-loxP system can be used for the modification of more than one gene, since this process occurs simultaneously, it is difficult to completely simulate the multi-step formation process of the tumor, and the mutation is a feature of the gradual accumulation formation process. Recently, researchers have used a dual-enzyme system that can function independently (such as Flp-FRT/Cre-loxP, or Cre-loxP/Dre-rox) to establish a modification of the target gene expression. The practical significance of the successful application of this technical method is as follows: 1. Independent research on spontaneous and non-spontaneous pathways and processes of tumor cells; 2. Simulating human multi-step cancer formation process, with continuous induction mutation; 3. Unique genetic evaluation of tumor treatment targets. Improvement of Tumor GEM Model Construction Strategy and Technology The GEM model has proven to be an effective tool for oncology research because of its unique advantages, but researchers have been working hard to continuously improve and improve it. Due to the long period of the construction and development process, the workload is high, and the cost is high, especially for the modification of multiple allelic genetic loci, this mouse model is constructed to construct a new mutant mouse model with genetic characteristics. It is more time consuming and requires a longer mating breeding process. This has also become a major factor limiting the widespread practical application of genetically capable GEM models. In recent years, due to the widespread popularization and application of tumor genome sequence research technology and the rapid growth of newly discovered cancer-related gene mutations, it is more necessary to establish a rapid and novel mouse model development strategy to achieve rapid verification of potential oncogenes in vivo. And the purpose of establishing a non-reproductive genetically modified GEM model of known patient-related mutations. At present, the progress in this area mainly includes: ES cell-based tumor model; application of CRISPR/Cas9 technology genome programming; and improved tumor model of tumor patient related sites. 1. ES cell-based tumor model In order to further accelerate the development of a novel human tumor GEM model, it has become a research strategy to directly perform oncology research on mouse embryonic stem cells (ESC) after genetic modification as non-genetically modified (eg, chimeric) mice. The recently reported GEM-ESC strategy is to establish an ES cell-based tumor GEM model. This kind of model is based on the original genetic modification, and rapidly constructs a novel genetically modified mouse model. For example, using the GEM-ESC strategy, the MET protooncogene was directly introduced into the ESC of the mouse breast tumor model of K14cre-Brca1-Trp53 (KBIP) to construct a novel breast cancer model of KBIP-MET. The results of the study showed that compared with KBIP mice, the breast cancer formed by this KBIP-MET mouse has more transformation characteristics, which is more likely to form carcinosarcoma. In response to tumor drugs, breast cancer in the KBIP mouse model is sensitive to clinical RARP inhibitors (such as Olaparib), while transformed breast tumors in the KBIP-MET mouse model show tolerance to the inhibitor. 2. Genomic programming using CRISPR/Cas9 technology In the past decade, with the rapid development of new genome programming techniques (such as ZFNs and TALENs), the CRISPR/Cas9 genome programming system appeared in 2013. This kind of genome editing technology has become the development of PCR technology in the past few years. Revolutionary and most influential technology updates. The CRISPR/Cas9 system was first discovered in prokaryotes against the immune system established against foreign invasive genetic material and was quickly used for gene editing of various species. With a single guide RNA (sgRNAs), Cas9 nucleases can specifically target any gene locus in the genome for gene knockout. By applying Cas9-induced DNA fragmentation and single-stranded nucleotide/donor DNA, the system can also be genetically modified for specific gene mutations, or for specific insertion of loxP/FRT recombination sites. The CRISPR/Cas9 technology system demonstrates the ability to efficiently edit gene-targeting strategies at different sites in the genome, making it the best choice for rapid development of tumor mouse models. All genetic mutations currently observed in human tumor patients can be rapidly constructed by genetic modification methods, including conditional gene knockouts, point mutations, translocations, and the like. In addition, researchers have used the CRISPR/Cas9 technology to perform somatic (non-reproductive hereditary) editing of mouse oncogenes and TSGs. Because of the efforts and success of this research strategy, the system has become a hepatocellular tumor, lung cancer, A new approach to brain cancer, pancreatic cancer, and non-genetically modified models of breast cancer. Recently, the CRISPR/Cas9 system has also been applied to genetic modification of target gene suppression (CRISPRi) or activation (CRISPRa). Such a modification system can be used to develop a corresponding oncogene, and/or to inhibit the induction and reversible activation of a TSGs gene in a mouse model. For example, with the CRISPRa-based system, the purpose of studying its carcinogenic potential is achieved by activating the transcription of oncogenes. Although the CRISPR/Cas9-based gene editing system has great potential, the system has certain defects in gene editing in vivo. For example, the current system strategy is not suitable for verifying the carcinogenic potential of potential oncogenes. In addition, the gene editing method of introducing Cas9 into somatic cells can cause a specific immune response of Cas9, resulting in the possibility that Cas9-expressing cells are cleared. In order to avoid these potential risks, it is possible to carry out corresponding experiments in immunodeficient mice, or to obtain a mouse model with immunological tolerance to Cas9 by genetic modification, and then carry out corresponding animal experiments. Finally, although it has been reported that the use of inducible Cas9n nickases that cause DNA single-strand breaks can reduce off-target effects, researchers must have a clear understanding of the application in order to completely avoid mediated by CRISPR/Cas9. Off-target mutations required for non-design are difficult. 3. Improved tumor model of tumor patient related sites It is necessary and practical to construct an ideal mutation model for tumor patients, to study the role of target genes in tumorigenesis, and to effectively evaluate drug effects. Because in human tumor suppressor genes (TSGs), many germ cell-dependent germ cell mutations and somatic mutations are missense or nonsense mutations, leading to the formation of mutant products or possibly functional truncated proteins. Such mutations are difficult to achieve by conditional knockout mouse models, because the conditional genetic modification strategy is to completely knock out one or several exons in the target gene to achieve the function of inactivating the target gene. Some studies have shown that a mouse mutation model constructed with reference to a TSG mutation associated with a tumor patient can produce a different phenotype than a complete knockout of the target gene. For example, compared with Trp53 knockout mice, patient-associated Trp53 hot-spot mutant mice showed more pronounced carcinogenic activity. Similarly, a conditional mouse model of the BRca1 gene mutation associated with BRCA1 breast cancer patients showed that breast tumors caused by mutations in specific RING regions of the Brac1 gene were more likely to be compared to the Brac1 complete knockout mouse model. Those drugs that destroy DNA are tolerant because their BRCA1 protein contains less RING activity. Studies have also shown that Brac1 protein contains less RING activity due to mutations, which appears to be more resistant to DNA-damaging drugs, and the results help to reveal the causal relationship between these mutations and the therapeutic response. Application range of GEM model in oncology research As a GEM model of primary tumorigenesis, it can be a systemic choice for in vivo analysis of interactions between cells themselves and cells during tumor formation, development, and tumor formation during metastasis. The human tumor GEM model has also been successfully applied to verify candidate drug targets, evaluate treatment effects, and evaluate drug tolerance mechanisms. Since the GEM model forms primary tumors in mice with intact immune systems, this model is more suitable for exploratory research in potential tumor immunotherapy. To establish a strategy and method for the close correlation between genetically modified mouse models and human diseases, and to provide a meaningful application platform for exploring and developing new methods and strategies for tumor therapy. It also provides information on clinical treatment effects and other information for the design and development of new anti-tumor treatments. The GEM model plays an important role in the research progress and contribution of tumor biology and transformational oncology in the following aspects. 1. Verify potential oncogenes On the basis of a large number of tumor sample sequencing studies to obtain increasing potential tumor genes, it is necessary and practical to establish a strategy for rapid verification of these potential tumor-associated genes in vivo. Given the speed and relative simplification factors, GEM-ESC and CRISPR/Cas9 technologies are the preferred method for quickly validating potential tumor genes. In particular, the use of somatic-based CRIPPR/Cas9-mediated gene editing technology to establish a non-genetically modified mouse model to achieve high-throughput in vivo verification of potential tumor genes. For example, a combination of DNA injection and in vivo electroporation is used to introduce 15 gRNAs/Cas9 expression plasmid mixtures of 13 different major tumor suppressor-related genes in pancreatic ductal adenocarcinoma (PDAC) into mature mice. In the pancreas, a mouse model in which these 13 genes were simultaneously modified was constructed. The results showed that more than 60% of these target genes in this PDAC mouse showed gene knockout, suggesting that CRISPR/Cas9-mediated mutations induced tumor formation. Similarly, the GEM model that induces Cas9 expression using Dox was also used to validate a variety of known intestinal tumor genes (such as Apc and Trp53). In addition to modifying TSGs, CRISPR/Cas9 technology is also used to verify the carcinogenicity of chromosomal rearrangements, such as the fusion of the Eml4-Alk gene observed in lung cancer patients. In addition, the use of the GEM model to validate candidate potential oncogenes from clinical patients and screened has also become a common strategy for studying tumor-related gene function. For example, Professor Hu Zhuowei's group recently studied the role of pseudokinase Trib3 in promoting the formation of acute promyelocytic leukemia (APL) by constructing conditional overexpression and knockout GEM models. The results showed that it was also in mouse bone marrow cells. Specific expression or knockdown of Trib3 and oncogenic protein PML-RARa (PR) fusion gene, Trib3 gene can significantly increase the role of PR-induced APL formation. Professor Lu Yi's group was the first to confirm the specific overexpression of Sry gene male/female mice in the liver tissue by constructing the GEM model of the Y chromosome sex determining region (Sry) gene-specific chemical carcinogen (DEN) Inducing the formation of hepatocellular carcinoma (HCC) in mice is more sensitive, suggesting that the Sry gene plays an important role in the formation of HCC. 2. Study the dependence of oncogenes Oncogene dependence refers to the fact that certain tumor formation is completely dependent on a single consensus oncogene. Since the modification of genes by the conditional GEM model is irreversible, it is not suitable for studying oncogene dependence. Therefore, it is necessary to select different regulatory induction strategies for corresponding research, such as the fusion of oncogenes and ERT to control their expression. It has been reported that the Trp53-ERT variant replaces the homozygous Trp53-established homozygous knock-in mouse, and the Trp53-ERT mouse induces Trp53 expression only in the presence of Tamoxifen, and has formed a tumor mouse model. On the basis of the study, the effect of re-purification of p53 function on existing tumors was studied. The results showed that on the basis of lymphoma caused by Eu-Myc, the recovery of Trp53 activity can produce rapid apoptosis and significantly increase the survival rate of mice. In addition, the reversible induction system of Doxycycline (Dox) regulating gene expression was also applied to the establishment of the GEM model, and the expression of human MYC proto-oncogene was induced by this system to cause tumor formation. After shutting down the expression of the MYC gene, the corresponding reaction of the formed tumor after the proto-oncogene was inactivated was observed. This study is a Tet-off induction system using Dox, which continuously expresses the human MYC transgene in mouse hematopoietic stem cells, induces the formation of malignant T-cell lymphoma and acute myeloid leukemia in mice, on the basis of which, if induced by the addition of Dox After stopping the expression of MYC, the tumor phenotype was also found to be weakened, and it was confirmed that this process was associated with tumor cell cycle death. The study also found that different tumor types are different for the long-term effects of stopping the activation of MYC expression in this reversible induction system. For example, transient inhibition of MYC expression in osteosarcoma, as sarcoma cells differentiate into mature bone cells, will continue to shrink sarcoma. On the contrary, although the inhibition of MYC expression causes diffuse atrophy of liver cancer, the remaining tumor cells are still in a latent state, and after re-opening MYC expression, their tumor characteristics can be quickly restored. 3. Crack the mechanism of spontaneous transfer formation Despite the ever-improving strategy of choice for cancer treatment, metastatic disease remains the leading cause of cancer deaths. The metastatic process is a complex multi-step process formed by the mutual interaction of tumor cells and the tumor microenvironment. The vast majority of preclinical metastasis studies in the past were performed using cell line inoculation models, and such models do not truly reflect the metastatic process in tumor patients. The GEM model can cause primary tumor development and metastasis formation, and is therefore an indispensable tool for studying the process of spontaneous metastasis formation of tumors that were unknown in the past. Due to the excessive growth of the primary tumor, the mice generally have to be sacrificed before the formation of a large range of metastases, which is a potential deficiency of the GEM model. This limitation can be solved by orthotopically transplanting GEM-derived tumor tissue, such as by surgical transplantation, to achieve the effect of intratumoral heterogeneity of the donor tumor, so that the metastasis process is close to the clinical common metastasis. disease. Some important findings have been obtained by using the GEM model to study the tumor metastasis process. Past studies have suggested that tumor metastasis occurs in the late stages of tumor formation. However, studies by the BALB-NeuT and MMTV-PyMT mouse breast tumor models have shown that early damage transfected cells have the ability to spread to bone marrow and lung tissue to form micrometastases. In addition, epithelial-to-mesenchymal cell metastasis (EMT) is thought to play a very important role in tumor cell transmission and metastasis. However, studies using pancreatic cancer and breast cancer GEM models have shown that tumor cells not only retain their epithelial cell characteristics, but also appear in metastatic lesions, suggesting that EMT is not necessary for tumor metastasis formation in these models. Furthermore, in exploring the complex relationship between tumor cells and the immune system during the process of tumor metastasis formation, the GEM model has clearly played a prominent role. For example, bone marrow immune cells, such as macrophages and neutrophils, play a crucial role in promoting the metastasis of different types of cancer. Recent studies have shown that breast tumors cause systemic inflammation, that is, the expansion of IL-17-derived T cells and secondary immunosuppressive neutrophils, which can trigger the spontaneous metastasis of the GEM model of lobular breast cancer, leading to GEM transplantation. Model of spontaneous metastasis of disease. The GEM model also plays an important role in revealing the involvement of related genes in inhibiting tumor metastasis. Recently, Liu Baohua's group applied Tet-ON to induce the expression of Sirt7 in GEM, revealing the mechanism of Sirt7 inhibiting the metastasis of primary pancreatic cancer. The results confirmed that Sirt7 induced by Dox significantly inhibited MMTV-PyMT mouse breast tumors. The role of lung metastasis, and its mechanism of action is achieved by regulating the TGF-β signaling pathway. Therefore, the GEM model plays an indispensable role in revealing the complexity of tumor metastasis and challenging the generally accepted theory that tumor metastasis is a metastatic process including advanced cancer cells including EMT. These important findings may provide an important reference for the treatment of patients with metastatic cancer. 4. Study the role of tumor microenvironment The GEM model has played an irreplaceable role in revealing the role of tumor cell external factors such as cancer-associated fibroblasts (CAFs) and immune cells such as cancer-associated fibroblasts (CAFs) and immune cells and tumor formation processes. CAFs regulate ECM and basement membrane formation by synthesizing extracellular matrix (ECM) components such as collagen, fibronectin, and laminin. Moreover, CAFs are a source of various soluble mediators, including matrix metalloproteinases (MMPs), which play an important role in promoting ECM transformation and enhancing their homeostasis in ECM. GEM model studies have shown that CAFs have a dual role in tumor formation. Using a K4-HPV6 squamous skin cancer mouse model, it was found that CAFs can stimulate tumor development by enhancing inflammation, angiogenesis, and ECM reconstitution during epithelial cell malignant transformation. In contrast, studies by two independent pancreatic cancer GEM models have shown that inhibition of CAFs in vivo has an effect of accelerating the process of tumor formation, suggesting a preventive effect of CAFs on tumors. Such contradictory phenomena are understandable for immune cells. Initially, immune cells are thought to be cells that can transform tumors by attack, inhibiting the process of tumor formation. However, recent studies have shown that these immune cells also have a function of promoting tumors. Studies of several different tumor types using mouse models have revealed a correlation between inflammation and tumors. For example, a mouse model of colitis-associated cancer, which specifically knocks out the NF-jB signaling system in myeloid immune cells, slows tumor growth in mice, indicating that it has a tumor-promoting effect. In addition, the K4-HPV6 mouse model study also showed that mast cells and bone marrow-derived cells play a role in promoting squamous skin cancer formation by activating MMP9 and re-adjusting the matrix structure. Applying the same skin cancer model, it is found that chronic inflammation has the effect of promoting neonatal tumor formation. So far, research has begun on the promotion of inflammation-induced tumor-associated macrophages and neutrophils. For example, based on the MMTV-PyMT breast cancer mouse model, knocking out an important macrophage-associated gene CSF1 (Colony-stimulating factor 1) revealed that the malignant process of breast tumors in this mouse was delayed. Similarly, inhibition of CXCR2, a chemokine that mediates neutrophil migration, has the effect of inhibiting intestinal tumor formation in APC mice. In summary, these studies emphasize the role of immune cells in the process of tumorigenesis and development. 5. Determine the source of tumor cells Revealing the source of cells during tumorigenesis will provide a very important theoretical basis for the development and improvement of therapeutic strategies. The application of the GEM model has successfully elucidated the cellular origin of certain different tumor types. In a small cell lung cancer (SCLC) study, Trp53 and Rb1 genes were expressed in Clara cells, neuroendocrine (NE) cells, and type II alveoli (SPC) by intratracheal injection of cell-specific Adeno-Cre viral vectors. Cells were specifically knocked out and analyzed for different times of tumorigenesis and tumor phenotype. The results indicate that NE cells are the main source of cells responsible for SCLC formation relative to SPC cells. In addition, cell-derived studies can provide unexpected results that are different from previous studies. For example, in the past BRCA1-based breast cancer research, the source cells of this type of cancer are basal epithelial stem cells. In the application of GEM model to BRCA1-induced basal-like breast cancer research, it is found that luminal progenitor cells are the true source of basal-like tumors. Recent studies from two different experiments have shown that genetic variation (such as the Pik3ca mutation) can significantly affect stem cell composition. Pik3ca point mutations (such as H047R) cause loss of the ability of mammary epithelial cells with lineage characteristics to differentiate into pluripotent stem-like states. Moreover, the cell source of Pik3ca breast tumors dominates the degree of malignancy, indicating that it is of great practical significance to accurately find the source of tumor cells in terms of improving the specificity of anticancer drugs and therapeutic effects. 6. Verify new drug targets Considering that not all oncogenes are necessary to maintain tumor formation, verify inactivated TSG or reduce oncogenes before the human clinical trials, ie, in preclinical animals, against the corresponding target drug. A test that can cause atrophy of an ongoing tumor becomes very important. The application of inducible mouse model can be used to verify the correlation of oncogene maintenance of tumors. For example, in the mouse model of breast cancer, the expression of oncogene Pik3ca is induced to induce atrophy of some tumors, suggesting that these tumors are "dependent". Sexually active P13K signals are closely related. However, most tumors eventually relapse due to an increase in Met or Myc, suggesting that these genetic lesions may induce tolerance to P13K inhibitors. This example demonstrates that the inducible GEM model can be applied to preclinical studies to achieve not only the goal of validating drug targets, but also to reveal the mechanism of drug tolerance formation. TSG is also likely to be an effective drug target. Loss of p53 function in tumors is caused by a dominant negative or inhibitory mutation of the p53 gene, as well as an increase/overexpression of its specific inhibitors MDM2 and MDM4. Genetic studies using a GEM model that reversibly inhibits p53 activity have shown that restoring p53 gene function can rapidly resolve established tumors, suggesting the development of inhibitory MDM2 molecules, thereby restoring p53 function, or restoring mutant p53 to wild The clinical significance of anti-tumor drugs with type-function p53. Similarly, the use of a GEM model that induces knockdown of APC to study colorectal cancer also suggests that induction of APC function can rapidly cause rapid and extensive tumor cell differentiation and sustained recurrence-free atrophy, which is an in vivo evaluation of APC/Wnt. The pathway serves as a therapeutic target for colorectal cancer caused by APC mutations. 7. Clarify therapeutic effects and tolerance In order to minimize the risk of failure of new anti-tumor therapies in clinical trials, it is more important to establish a pre-clinical and predictive in vivo model to objectively assess the corresponding drug effects and tolerability. A study of the GEM model of lung cancer and pancreatic cancer caused by the Kras mutation found that the response effect of the GEM model on targeted therapy and conventional chemotherapy is very similar to that of the corresponding patient.需è¦åŠ 以关注的是å°é¼ 与人在è¯ç‰©ä»£è°¢æ–¹é¢è¡¨çŽ°æ˜Žæ˜¾ä¸åŒï¼Œæ¯”如,å‚与è‚è„è¯ç‰©ä»£è°¢çš„ç»†èƒžè‰²ç´ P450酶底物特异性方é¢ï¼Œä¸åŒçš„物ç§å˜åœ¨è¾ƒå¤§çš„差异。该类问题å¯å€ŸåŠ©äººæºåŒ–å°é¼ æ¨¡åž‹åŠ ä»¥è§£å†³ã€‚å› æ¤ï¼Œå»ºç«‹äººæºåŒ–çš„GEM模型作为临床å‰è¯ç‰©æ•ˆæžœçš„ç ”ç©¶ï¼Œå°†æœ‰åŠ©äºŽä¼˜åŒ–é’ˆå¯¹é¶ç‰¹å¼‚抗肿瘤è¯ç‰©çš„ç ”å‘,以åŠå¯»æ‰¾ä¸Žç¡®å®šæ²»ç–—æ•ˆåº”çš„å…³é”®å› ç´ ï¼Œå¹¶ä½¿å…¶æˆä¸ºè‚¿ç˜¤ç—…人特å¾çš„é¢„æµ‹æ€§ç”Ÿç‰©æ ‡è®°ã€‚å¦å¤–,GEM模型也å¯åº”用于探索治疗æ•æ„Ÿæ€§è‚¿ç˜¤èŽ·å¾—性è€è¯çš„å½¢æˆæœºåˆ¶ã€‚ 在探讨肿瘤治疗效应与è€å—机制方é¢ï¼ŒK14cre; Brca1-f/f; Trp53-f/f (KB1P) å°é¼ 是作为BRCA1çªå˜ä¹³è…ºç™Œä¸´åºŠå‰GEM模型的一个éžå¸¸æœ‰è¯´æœåŠ›çš„例å。KB1På°é¼ å¯å½¢æˆå®Œå…¨æ¨¡æ‹Ÿç±»äººBRCA1çªå˜ä¹³è…ºç™Œç»„织病ç†å¦ç‰¹å¾çš„乳腺肿瘤,而且,对å«é“‚ç±»è¯ç‰©å’ŒPARP抑制剂也具有高æ•æ„Ÿæ€§ã€‚临床试验è¯å®žï¼ŒPARP抑制剂Olaparibå¯ä»¥æ²»ç–—åµå·¢è‚¿ç™Œï¼Œä¹³è…ºç™Œå’Œç»“ç›´è‚ ç™Œã€‚ 虽然该è¯ç‰©å¹¶ä¸å¯¹æ‰€æœ‰è¿™ç±»ç™Œç—‡ç—…人的治疗都有效果,但其对BRCA1çªå˜æºå¸¦è€…表现有明显治疗效果,å¯èƒ½ä¸ŽPARP抑制剂åˆç”¨å¼•èµ·çš„ååŒè‡´æ»ä½œç”¨ä¸ŽBRCA1缺ä¹æœ‰å…³ã€‚BRCA1çªå˜ç»†èƒžå¯¹PARPæŠ‘åˆ¶å‰‚è¡¨çŽ°ä¸ºæ›´åŠ å®¹æ˜“è¢«æŸä¼¤ï¼Œå› 为PARP抑制剂诱å‘çš„å•ä¸€é“¾DNAæ–裂,å¯å¯¼è‡´DNAå¤åˆ¶æ—¶çš„åŒé“¾æ–è£‚ï¼Œé€ æˆBRCA1ç¼ºå¤±ç»†èƒžæ— æ³•å®žæ–½åŒæºé‡ç»„机制æ¥ä¿®å¤æŸä¼¤çš„DNA。 æ ¹æ®Olaparib在临床试验ä¸èŽ·å¾—çš„ç†æƒ³æ•ˆæžœï¼ŒFDA于2014å¹´12月批准了该è¯ç”¨äºŽBRCA1/2çªå˜åµå·¢ç™Œç—…人的治疗。尽管病人对该è¯ç‰©æœ‰å¾ˆå¥½çš„å应效果,然而,在病人和GEMæ¨¡åž‹ç ”ç©¶ä¸éƒ½å‘现了获得性è¯ç‰©è€å—性。通过临床å‰KB1På°é¼ æ¨¡åž‹ç ”ç©¶è¯å®žï¼Œè¿™ç±»è€è¯æœºåˆ¶ä¸Žè¯ç‰©è¿è¾“物åŠåŒæºé‡ç»„æ¢å¤ç‰æ•°é‡çš„å¢žåŠ æœ‰å…³ã€‚è¿™äº›ç ”ç©¶ç»“æžœæœ‰åŠ©äºŽäº†è§£ä¸´åºŠä¸Šè€è¯æ€§çš„产生, 以åŠè®¾è®¡é’ˆå¯¹Olaparibè€è¯ç—…人的改进治疗ç–略。 关于肿瘤治疗效应与è¯ç‰©è€å—性的关系,现在也是越æ¥è¶Šæ¸…楚,å³å…¶å½±å“ä¸ä»…å—è‚¿ç˜¤ç»†èƒžè‡ªèº«å› ç´ ï¼Œè€Œä¸”ä¸Žæˆçº¤ç»´ç»†èƒžå’Œå…疫细胞ç‰åŸºè´¨å› ç´ æœ‰å…³ã€‚é€šè¿‡PDACçš„GEMæ¨¡åž‹è¿›è¡Œè‚¿ç˜¤å¹²é¢„ç ”ç©¶çš„ç»“æžœè¡¨æ˜Žï¼Œæ²»ç–—æ˜¯æŠ‘åˆ¶æ—泌性相关信å·é€šè·¯ï¼Œå‡å°‘ä¿ƒç»“ç¼”ç»„ç»‡å¢žç”Ÿè‚¿ç˜¤åŸºè´¨ï¼Œå¢žåŠ è‚¿ç˜¤è„‰ç®¡ç³»ç»Ÿï¼Œå¯¼è‡´ä¿ƒè¿›æŠ—è‚¿ç˜¤è¯ç‰©å¯¼å…¥è‚¿ç˜¤éƒ¨ä½çš„过程。然而,关于攻击PDACä¸çš„è‚¿ç˜¤åŸºè´¨çš„æ¦‚å¿µï¼Œæœ€è¿‘æœ‰ä¸¤ä¸ªç ”ç©¶å¯¹è¯¥ç†è®ºè¿›è¡Œäº†æŒ‘æˆ˜ã€‚è¿™ä¸¤ç ”ç©¶ç»“æžœè¡¨æ˜Žï¼ŒåŸºè´¨å› ç´ å¯èƒ½é€šè¿‡é˜»æ¢è‚¿ç˜¤è¡€ç®¡å½¢æˆï¼Œå¼•èµ·ä¸æ˜¯ä¿ƒè¿›è€Œæ˜¯æŠ‘制PDACç”Ÿé•¿ã€‚æ‰€ä»¥ï¼Œè¿™äº›ç ”ç©¶éƒ½æ示了肿瘤微环境在治疗è€å—性方é¢ï¼Œå‘æŒ¥äº†æ¯”è¿‡åŽ»æƒ³è±¡çš„æ›´åŠ é‡è¦çš„,且å¤æ‚的作用。 8. 肿瘤å…疫治疗 在过去å年,通过对å…ç–«å应的深刻æ示与认识,建立了利用病人å…疫系统攻击肿瘤的治疗ç–略。近年æ¥çš„é»‘è‰²ç´ ç˜¤å’Œè‚ºç™Œä¸´åºŠç—…äººè¯•éªŒå·²è¯å®žï¼ŒåŒ…括抗CTLA-4和抗PD-1çš„å…疫检查点抑制物,在增强病人有效抗肿瘤å…ç–«åŠæ”¹å–„生å˜çŽ‡ç‰æ–¹é¢å…·æœ‰å·¨å¤§æ½œåŠ›ã€‚这些临床试验基础是æ¥è‡ªè¿‡åŽ»å‡ å年在实验å°é¼ æ¨¡åž‹ä¸Šå¼€å±•çš„åŸºç¡€ç ”ç©¶ï¼Œä»Žè€Œæ示了CTLA-4å’ŒPD-1在阻æ¢å…ç–«å应ä¸çš„é‡è¦æ€§ï¼Œç‰¹åˆ«æ˜¯é€šè¿‡å¯¹CTLA-4å’ŒPD-1敲除å°é¼ 出现严é‡ä¸Žæ¸©å’Œç¨‹åº¦çš„自å‘性自身å…疫表型的明确è¯å®žï¼Œåº”用CTLA-4抑制物引起接ç§è‚¿ç˜¤å°é¼ 的抗肿瘤T细胞ååº”å¢žåŠ ï¼Œäº§ç”Ÿè‚¿ç˜¤æŽ’æ–¥ä½œç”¨ï¼Œæ示释放T细胞上的刹车å¯èƒ½æ˜¯æŠµæŠ—肿瘤的一个潜力的应对ç–略。尽管如æ¤ï¼ŒåŒæ—¶ä¹Ÿåº”该清楚的了解,ä»æœ‰ä¸€å®šæ¯”例的病人对æ¤ç±»å…疫治疗没有å应,且目å‰çš„挑战是还ä¸çŸ¥é“其真æ£åŽŸå› 。 ç›®å‰ï¼Œè™½ç„¶ç»å¤§éƒ¨åˆ†çš„å…ç–«å¦ç ”究都是在肿瘤移æ¤å°é¼ æ¨¡åž‹çš„åŸºç¡€ä¸Šè¿›è¡Œçš„ï¼Œä½†å°±çŽ°åœ¨ç ”ç©¶æƒ…å†µé¢„æµ‹è¡¨æ˜Žï¼Œä»ŠåŽGEMæ¨¡åž‹åº”ç”¨äºŽè¯¥é¢†åŸŸçš„ç ”ç©¶å°†ä¼šè¶Šæ¥è¶Šå¤šã€‚应用GEMæ¨¡åž‹ç ”ç©¶çš„éƒ¨åˆ†ç»“æžœè¡¨æ˜Žï¼Œåœ¨æ–°å½¢æˆçš„肿瘤过程ä¸ï¼Œå› 肿瘤引起的è€å—机制,T细胞丧失其对肿瘤细胞的å应性,特别明显的是,如果将æ¥è‡ªGEM模型å°é¼ å½¢æˆçš„肿瘤细胞,接ç§è‡³å…疫缺陷å°é¼ 体内,肿瘤会快速生长,而野生å°é¼ 则能排斥这些肿瘤细胞。æ示这些肿瘤细胞没有失去其å…疫原性,T细胞ä»ç„¶èƒ½è¯†åˆ«è¿™äº›ç»†èƒžå¹¶å‘挥其攻击作用。但在原å‘肿瘤å°é¼ 的体内这些T细胞å´æ— 能为力。 肿瘤常常被认为是慢性炎症的结果,这ç§ç‚Žç—‡å¯å¼•èµ·å±€éƒ¨å’Œç³»ç»Ÿå…疫抑制,从而ä¸åˆ©äºŽT细胞å‘æŒ¥å…¶æœ‰æ•ˆçš„åŠŸèƒ½ã€‚è€Œä¸”ï¼Œè‚¿ç˜¤å¸¸è¡¨çŽ°ä¸ºæ ‘çŠ¶çªç»†èƒžï¼ˆDC)功能缺失,导致T细胞å¯åŠ¨ç¼ºæŸã€‚例如,在MMTV-PyMT乳腺肿瘤å°é¼ 模型ä¸å‘现,原本具有潜在激活抗肿瘤T细胞的DC细胞,会被大é‡å˜åœ¨çš„巨噬细胞竞争抑制,这些巨噬细胞起到了阻æ¢ç‰¹å®šTç»†èƒžæ¿€æ´»çš„ä½œç”¨ã€‚æœ€è¿‘çš„ç ”ç©¶ä¹Ÿè¯å®žï¼Œä¿ƒè¿›DC细胞功能或阻æ¢éª¨é«“细胞引起的å…疫抑制,å¯ä»¥è¾¾åˆ°æ”¹å–„å…ç–«æ£€æŸ¥ç‚¹æŠ‘åˆ¶ç‰©çš„æŠ—è‚¿ç˜¤æ•ˆæžœã€‚å› æ¤ï¼Œå¯¹è‚¿ç˜¤ç—…人实施è€å—T细胞å…疫激活疗法的时候,结åˆåº”用针对改善å…疫抑制或则增强T细胞å¯åŠ¨çš„é¶ç‚¹è¯ç‰©ï¼Œåœ¨ä¸´åºŠä¸Šå¯èƒ½ä¼šèµ·åˆ°æ›´å¥½çš„治疗效果。 相对于肿瘤接ç§æ¨¡åž‹ï¼Œåº”用GEM模型进行å…ç–«æ²»ç–—ç ”ç©¶éœ€è¦ä¸åŒçš„方法。考虑到GEM模型ä¸è‚¿ç˜¤æ˜¯åœ¨æ¯ä¸ªç‹¬ç«‹å°é¼ 体内å‘生的,如åŒç—…人肿瘤的å‘ç”Ÿè¿‡ç¨‹ï¼Œå…·æœ‰ç‰¹æ®Šçš„è‚¿ç˜¤æŠ—åŽŸã€‚å› è€Œï¼Œå…·æœ‰å…¶å¼‚è´¨æ€§çš„ç‰¹ç‚¹ï¼Œä»Žè€Œç¡®ä¿åŒºåˆ«é‰´å®šå应和éžå应肿瘤之间的分åä¸åŒï¼Œä¹Ÿæœ‰åŠ©äºŽä¸´åºŠé¢„æµ‹çš„ç”Ÿç‰©æ ‡è®°ç‰©çš„å»ºç«‹ã€‚ç„¶è€Œï¼Œå¯¹äºŽå¤§å¤šæ•°GEM模型æ¥æºçš„肿瘤æ¥è¯´ï¼Œè¾¨è®¤å¯è¢«T细胞识别的表达肿瘤抗原å´æ˜¯æœªçŸ¥çš„。为了克æœè¿™ç‚¹ï¼Œå¯å€ŸåŠ©åŸºå› 修饰的方å¼ï¼Œå°†ä¸´åºŠä¸Šç›¸å…³è‚¿ç˜¤æŠ—原引入å°é¼ 体内,使其具有诱å‘肿瘤特异性T细胞å应的效果。比如,应用低å…疫原性的肿瘤(如肉瘤和肺癌),将肿瘤特异性抗原引入至GEM模型åŽï¼Œè¿™äº›è‚¿ç˜¤çš„å…ç–«åŽŸæ€§å¢žåŠ ï¼Œå¼•èµ·æ½œåœ¨è€ŒçŸæ—¶é—´çš„抗肿瘤T细胞å应。起åˆçš„抗肿瘤T细胞å应很快å‘生,然åŽæ˜¯å¯è°ƒæŽ§T细胞介导的å…ç–«æŠ‘åˆ¶ã€‚å› æ¤ï¼Œè¿™äº›æ¨¡åž‹å°†æœ‰åŠ©äºŽç›®å‰ä¸Žæœªæ¥ç›¸å…³ç ”究,以达到æ示å…疫逃逸的å¤æ‚æœºåˆ¶ï¼Œæœ€ç»ˆç ”å‘具有改善的肿瘤å…疫治疗的新ç–略的目的。 9. 与临床试验并行的 GEM 模型 最近推出的“与临床共试验†范例,目的是将临床å‰GEM实验与人体临床试验åŒæ—¶å¼€å±•ï¼Œä»Žè€Œè¾¾åˆ°é¢„测治疗效果的作用。该ç–略已ç»åº”用于å‰åˆ—腺癌治疗ä¸ï¼Œå¹¶æˆåŠŸæç¤ºäº†ç”±é›„æ€§æ¿€ç´ è¯±å‘çš„è€å—性与æŸäº›é—ä¼ å…³é”®å› å有关,以åŠå…‹æœåŽ»åŠ¿éš¾æ²»æ€§çš„新综åˆç–—法。åŒæ ·ï¼Œåº”用NSCLCçš„GEM模型的一起临床试验表明,Kras/Lkb1çªå˜è‚ºç™Œè¾ƒKras or Kras/p53çªå˜è‚¿ç˜¤å¯¹ä¸´åºŠä¸Šçš„Docetaxelå’ŒMEK抑制剂Selumetinibçš„è”åˆç–—æ³•ï¼Œè¡¨çŽ°ä¸ºæ›´åŠ å…·æœ‰è€å—性。æ示了LKB1是临床试验ä¸å¯¹æ¤ç±»è¯ç‰©è”åˆç–—法è€å—çš„æ½œåœ¨å†³å®šå› ç´ ã€‚è¿™ç±»ç ”ç©¶è¡¨æ˜Žï¼Œåº”ç”¨GEM模型作为人肿瘤临床å‰è¯ç‰©æ•ˆæžœç ”究,能å‘çŽ°æ–°çš„ç”Ÿç‰©æ ‡è®°ç‰©å’Œè”åˆç–—法。 GEM在肿瘤å¦ç ”究ä¸å‘展趋势与未æ¥å±•æœ› 许多抗肿瘤è¯ç‰©åœ¨ä¸´åºŠè¯•éªŒä¸æœªèƒ½è¾¾åˆ°ä¸´åºŠå‰å®žéªŒçš„期望目的,已ç»æˆä¸ºç›®å‰è‚¿ç˜¤å¦ä¸Žè½¬åŒ–癌症医å¦æ‰€é¢ä¸´çš„巨大挑战,如何改善肿瘤å¦é¢†åŸŸçš„临床å‰ç ”究结果的预测性,也是人们å分关注的çƒç‚¹ã€‚å› æ¤ï¼Œå¦‚何选择更能真实åæ˜ äººè‚¿ç˜¤ç–¾ç—…å‘生å‘展过程的临床å‰è‚¿ç˜¤åŠ¨ç‰©æ¨¡åž‹ï¼Œå°†æ˜¾å¾—æ›´åŠ é‡è¦äº†ã€‚为了实现这一目的,首先需è¦è€ƒè™‘的是如何建立能真实åæ˜ è‚¿ç˜¤æœ¬èº«åŠå¤–在特å¾çš„临床å‰å°é¼ 模型。比如,临床å‰æ¨¡åž‹åº”该å«æœ‰ç—…人特异性çªå˜ï¼Œè¯¥ç§çªå˜å…·æœ‰è¯±å‘æ¶æ€§è‚¿ç˜¤çš„趋势,且在一定病人群ä¸æ˜¾ç¤ºé—ä¼ å˜åŒ–特å¾ã€‚å¦å¤–,原å‘的肿瘤过程是在自然微环境ä¸è¿›è¡Œçš„,如åŒåœ¨äººè‚¿ç˜¤è¿›è¡Œè¿‡ç¨‹ä¸æ‰€è§çš„肿瘤细胞与肿瘤微环境之间相互作用(包括å…疫细胞,æˆçº¤ç»´ç»†èƒžï¼Œä»¥åŠæ·‹å·´ç»†èƒžå’Œè¡€ç®¡çš„潜入)。å†æœ‰ï¼Œç”±äºŽç»å¤§å¤šæ•°è¿›å…¥ä¸´åºŠè¯•éªŒçš„病人已ç»æ˜¯å¤„于广泛肿瘤转移疾病过程ä¸ï¼Œå› æ¤ï¼Œå®žéªŒè®¾è®¡æ—¶å°±åº”该考虑选择能模拟病人疾病ä¸åŒè¿›ç¨‹çŠ¶æ€çš„å°é¼ 模型,开展相应的临床å‰çš„è¯ç‰©æ•ˆæžœè¯„ä¼°ç ”ç©¶ã€‚åœ¨çŽ°å®žçš„ä¸´åºŠè¯•éªŒä¸ï¼Œå¾€å¾€è¿›å…¥ä¸´åºŠè¯•éªŒçš„病人之å‰å¤šæ˜¯ç»è¿‡ä¸åŒç¨‹åº¦çš„æ²»ç–—ï¼Œå› è€Œæžæœ‰å¯èƒ½ä¼šå¹²æ‰°æ²»ç–—效果的验è¯ã€‚而临床å‰çš„åŠ¨ç‰©ç ”ç©¶åˆ™æ˜¯å»ºç«‹åœ¨ä»Žæ¥æ²¡æœ‰æŽ¥å—任何治疗的基础上进行的,从而导致高估治疗效果的结果。从å¦å¤–一方é¢è®²ï¼Œä¸´åºŠè¯•éªŒä¸å¯¹é‚£äº›æŽ¥å—过治疗的严é‡ç—…人没有效果,å´ä»æœ‰å¯èƒ½å¯¹æ²¡æœ‰æŽ¥å—过治疗的严é‡ç—…人有益处。 æœ€è¿‘åŸºå› ä¿®é¥°æ¨¡å¼åŠ¨ç‰©æŠ€æœ¯çš„è¿›å±•ï¼Œä¿ƒè¿›äº†å¿«é€Ÿç ”åˆ¶æ›´åŠ ç²¾å‡†çš„èƒ½å¼•èµ·åŽŸå‘肿瘤的å°é¼ 模型,该类模型结åˆäº†ç‰¹å¼‚性肿瘤病人形æˆå‘生å‘展过程ä¸è‚¿ç˜¤ç»†èƒžæœ¬èº«å’Œç»†èƒžå¤–的特å¾ã€‚预测这些新一代的GEM模型和基于GEM模型的移æ¤æ¨¡åž‹ï¼Œå°†æ˜¯çœŸå®žæ¨¡æ‹Ÿç—…人肿瘤å‘生å‘å±•è¿‡ç¨‹ï¼Œç ”ç©¶è‡ªå‘性转移疾病的最佳模å¼åŠ¨ç‰©ã€‚这些模型å¯ä½œä¸ºç ”究肿瘤形æˆçš„å¤æ‚过程(包括肿瘤的起始,器官特异性转移的形æˆï¼Œè‚¿ç˜¤å¾®çŽ¯å¢ƒçš„å‚与ç‰æ–¹é¢ï¼‰çš„é‡è¦è€Œæœ‰ç”¨å·¥å…·ã€‚ä½†æ˜¯ï¼Œå¯¹äºŽè‚¿ç˜¤ç—…äººè€Œè¨€ï¼Œæ›´åŠ é‡è¦çš„是,这些模型能更为深入地æ示å…疫治疗ä¸çš„å应性与è€å—性,以åŠç–¾ç—…çš„å¤å‘ç‰ç›¸å…³æœºåˆ¶ã€‚期待未æ¥ï¼Œåº”用新一代GEM模型对抗肿瘤新è¯ç‰©ä¸´åºŠå‰çš„è¯„ä¼°ç ”ç©¶ï¼Œå°†ä¼šå¢žåŠ é¢„æµ‹å…¶åœ¨ä¸´åºŠè¯•éªŒä¸çš„æˆåŠŸçŽ‡ï¼Œä»Žè€ŒåŠ 速抗肿瘤新è¯ç–略设计与临床实施,达到改善防治肿瘤病人的病情的最终目的。 About the Author 俞晓峰åšå£«ï¼Œå›½é™…知å模å¼åŠ¨ç‰©å’Œç»†èƒžç”Ÿç‰©å¦ä¸“家,先åŽå°±ä»»äºŽè€¶é²å¤§å¦åŒ»å¦é™¢ã€iTLåŸºå› æ‰“é¶å…¬å¸å’Œçº½çº¦å¤§å¦åŒ»å¦é™¢ä»¥åŠç¾Žå›½ASC生物技术公å¸ç‰æœºæž„,在é—ä¼ ä¿®é¥°æ¨¡å¼åŠ¨ç‰©é¢†åŸŸæœ‰è¶…过20å¹´çš„ç ”å‘和管ç†ç»éªŒã€‚ç›®å‰ä»»èŒäºŽèµ›ä¸šç”Ÿç‰©ç§‘技,任高级副总è£å’Œé«˜çº§ç§‘å¦å®¶ï¼Œä¸»è¦è´Ÿè´£åŸºå› 修饰模å¼åŠ¨ç‰©å¹³å°çš„æŠ€æœ¯å·¥ä½œï¼Œå…¶ç ”ç©¶æˆæžœå¤šæ¬¡å‘表在Nat Immunol 〠Mol Cell Biolç‰é«˜æ°´å¹³æ‚志上。 主è¦å‚考文献: 1. Li K, Wang F,Cao WB, and ZW et al (2017) TRIB3 Promotes APL Progression through Stabilization of the Oncoprotein PML-RARa and Inhibition of p53-Mediated Senescence. Cancer Cell 31, 697–710 2. Tang X,Shi L,and Liu B et al (2017) SIRT7 antagonizes TGF-β signaling and inhibits breast cancer metastasis. Nature Communications 8: 318 3. Liu C, Ren YF, Yi Lv and Xu-Feng Zhan et al (2017) Activation of SRY Accounts for Male-Specific Hepatocarcinogenesis: Implication in Gender Disparity of Hepatocellular Carcinoma. Cancer Letters 410: 20-31 4. Chunga WJ, Daemena A, Melissa R, and Junttila MR et al (2017) Kras mutant genetically engineered mouse models of human cancers are genomically heterogeneous. PNAS Dec 4, E10947–E10955 5. Annunziato S, Kas SM, Nethe M, and Drenth AP et al (2016) Modeling invasive lobular breast carcinoma by CRISPR/Cas9-mediated somatic genome editing of the mammary gland. Genes Dev 30: 1470 – 1480 6. Drost R, Dhillon KK, and Schut E et al (2016) BRCA1185delAG tumors may acquire therapy resistance through expression of RING-less BRCA1. J Clin Invest 126: 2903 – 2918 7. Maresch R, Mueller S, and Barenboim M et al (2016) Multiplexed pancreatic genome engineering and cancer induction by transfection-based CRISPR/ Cas9 delivery in mice. Nat Commun 7: 10770 8. Chiou SH, Winters IP, Wang J, and Chuang CH et al (2015) Pancreatic cancer model-ing using retrograde viral vector delivery and in vivo CRISPR/Cas9-mediated somatic genome editing. Genes Dev 29: 1576 – 1585 9. Weber J, Öllinger R, and Engleitner T et al (2015) CRISPR/Cas9 somatic multiplex-mutagenesis for high-throughput functional cancer genomics in mice. Proc Natl Acad Sci USA 112: 13982 – 13987 10. Dow LE, O'Rourke KP, and Lowe SW et al (2015b) Apc restoration promotes cellu-lar differentiation and reestablishes Crypt homeostasis in colorectal cancer. Cell 161:1539 – 1552 11. Henneman L, van Miltenburg MH, and Schlicker A et al (2015) Selective resistance to the PARP inhibitor olaparib in a mouse model for BRCA1-deficient metaplastic breast cancer. Proc Natl Acad Sci USA 112: 8409 – 8414 12. Clohessy JG, Pandolfi PP (2015) Mouse hospital and co-clinical trial project- from bench to bedside. Nat Rev Clin Oncol 12: 491 – 498 13. Cong L, Ran FA, Cox D, and Marraffini LA et al (2013) Multiplex genome engineering using CRISPR/Cas systems. Science 339: 819 – 823 14. Jamieson T, Clarke M, and Nibbs RJB et al (2012) Inhibition of CXCR2 profoundly suppresses inflammation-driven and spontaneous tumorigenesis. J Clin Invest 122: 3127 – 3144 15. Drake AC, Chen Q, Chen J (2012) Engineering humanized mice for improved hematopoietic reconstitution. Cell Mol Immunol 9: 215 – 224 16. Liu P, Cheng H, and Fox EA et al (2011) Oncogenic PIK3CA-driven mammary tu-mors frequently recur via PI3K pathway-dependent and PI3K pathway- independent mechanisms. Nat Med 17: 1116 – 1120 17. Martins CP, Brown-Swigart L, and Evan GI (2006) Modeling the therapeutic efficacy of p53 restoration in tumors. Cell 127: 1323 – 1334 18. Shibata H, Toyama K, Shioya H, and Toyoshima K et al (1997) Rapid colorectal adenoma formation initiated by conditional targeting of the Apc gene. Science 278: 120 – 123
We are Hospital Electric From China Manufacturer Medical Bed, also known as Hospital Bed Home Care, Hospital Equipment, Nursing Bed, etc., is a hospital bed used by patients when they are hospitalized in the hospital.
There are many classifications of medical beds, and the specific classification methods are as follows: According to the material, it can be divided into ABS medical beds, all stainless steel medical beds, semi-stainless steel medical beds, all steel sprayed medical beds, etc.
According to the function, it can be divided into electric medical beds and manual medical beds. Among them, electric medical beds can be divided into five-function electric medical beds and three-function electric medical beds. Manual medical beds can be divided into double-shake medical beds and single-shake medical beds. Medical beds, flat medical beds.
According to whether it can be moved, it can be divided into a wheeled medical bed and a right-angle medical bed. Among them, electric medical beds are generally movable with wheels.
Hospital Bed Mattress,Anti Bedsore Air Mattress,Inflatable Air Mattress,Homecare Bed Mattress Haloxylon Ammodendron Medical Equipment Co., Ltd. , https://www.ssmedicaldevic.com