Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2022 Nov 1.
Published in final edited form as: Curr Protoc. 2021 Nov;1(11):e284. doi: 10.1002/cpz1.284

An Overview of Preclinical Solid Tumor Models to Study Novel Therapeutics in Brain Metastases

Mohini Singh 1, Ashish Dahal 1, Priscilla K Brastianos 1
PMCID: PMC8597918  NIHMSID: NIHMS1743336  PMID: 34762346

Abstract

Metastases are the most common malignancy of the adult central nervous system and are becoming an increasingly troubling problem in oncology largely due to the lack of successful therapeutic options. The limited selection of treatments is a result of the currently poor understanding behind the biological mechanisms of metastatic development, which in turn is difficult to achieve because of limited preclinical models that can accurately represent the clinical progression of metastasis. Described in this unit are in vitro and in vivo model systems that are used to enhance the understanding of metastasis, and for identifying new therapies for the treatment of brain metastasis.

Keywords: Brain metastasis, preclinical models, in vitro models

Introduction

Metastases are secondary malignancies that form when cancer cells break away from a primary tumor and spread to other locations within the body (1). Of all organs for these escaped tumor cells to colonize, the brain accounts for an estimated 9–17% of cases in patients with systemic cancer (2), though the exact incidence varies with several factors, such as primary site, staging, and even subtype. Brain metastases are the most frequently occurring tumor type of the adult central nervous system (CNS), being 10-fold greater than primary malignant brain tumors (3, 4). The frequency of brain metastases development varies with the primary cancer, occur most frequently with lung (20%), melanoma (7%), and breast (5%) primary cancers (5). As treatments for primary cancers improve, metastases are quickly becoming the major cause of morbidity and mortality in patients with solid cancers (1, 6). This is due in part to improved control of the primary tumors, resulting in longer survival times and subsequently more time for brain metastases to develop, as well as enhanced detection methods (7). While limited treatment options are available for survival of brain metastasis patients beyond a few months (5, 7), groundbreaking therapeutic modalities for various patient subsets are improving control and overall prognosis of brain metastasis (810). Several target-specific therapies have been developed for brain metastasis possessing various molecular alterations, such as EGFR (11), HER-2 (12), and BRAF (13, 14). Although meaningful extensions of survival are being achieved, only a subset of patients respond to therapy, and many of these therapies are associated with limiting side effects.

The mechanisms regulating brain metastasis initiation remain elusive, partly due to the inability to identify early stages of metastatic progression with current detection and visualizing techniques, thereby limiting, the development of preventative therapies (15). It is therefore essential to have in vitro and in vivo models that can recapitulate clinical disease progression to gain insights into the metastatic process and for identifying new therapeutic targets.

The Metastatic Process

Metastasis occurs when cancer cells successfully migrate from a primary tumor to secondary sites in the body. The metastatic process follows a sequence of well-defined steps that allow cancer cells to grow beyond their primary tumor site (16, 17). To detach from the primary tumor mass, the cancer cells first downregulate intercellular adhesion molecules such as cadherins and integrins (18). Normal cells that detach from their surrounding tissue typically undergo cell death via anoikis (19). However, migrating cancer cells ensure their survival by auto-activating survival signaling pathways such as FAK and the PI3k/Akt (20). Without requiring anchorage to surrounding cells or the extracellular matrix, the migrating cancer cells can continue traveling towards vasculature.

Upon reaching blood or lymph vessels, migrating cancer cells intravasate past endothelial cells and enter the systemic circulation (21). They must avoid immune detection or risk elimination. Metastasizing cancer cells employ a variety of strategies, from recruiting immunosuppressive cell populations to hijacking the function of normal immune cells (22). Upon successfully reaching distant sites, the migrating cancer cells extravasate from the circulatory system and colonize into satellite tumors (23). Some cancers subtypes are particularly suited for particular secondary metastasis sites, a phenomenon known as the seed-soil hypothesis (24).

Blood Brain Barrier

For brain metastasis to occur, the migrating cancer cells must penetrate the blood-brain barrier (BBB), a semipermeable border in the brain’s blood vessels that tightly regulates the movement of cells and solutes from the bloodstream into the CNS (25). The BBB is primarily composed of endothelial cells, mural cells, basement membrane, and astrocytes that form a network to prevent pathogens and toxins from infiltrating neural tissue (25). However, this barrier also prevents most circulating immune cells from entering the brain (26). Metastasizing cancer cells increase leakage of the BBB by disrupting endothelial cell transporters such as Mfsd2a (27) and once inside the brain, cancer cells utilize the BBB to shield themselves from immune surveillance (26). Although the exact mechanisms regulating cancer cell infiltration of the BBB is unknown, the compromised BBB is restructured into what is referred to as the blood-tumor barrier (BTB) (28). The BTB contains heterogenous vasculature that actively nurtures metastasized cells before they develop their own blood vessel networks in the brain (29). The dual role of the BBB in protecting metastasized cancer cells from the immune system and preventing drugs from entering the brain makes treatment of brain metastases particularly challenging (30).

The exact mechanisms regulating the metastatic process remain elusive, partly due to the inability to identify the early stages of metastatic progression with available detection and visualizing techniques (31). The development of assays and models that recapitulate the features of clinical disease progression is a way to address this issue.

In vitro models of metastasis

An ideal in vitro assay should mimic at least one of the following features of the metastatic cell in vivo: proliferation, migration/invasion and morphological characteristics, cellular and phenotypic heterogeneity, microenvironment and drug response (Table 1) (41). However, despite the fact that complexity of the metastatic cascade makes it difficult to replicate each step precisely, a number of assays have been developed that have proven highly valuable. Cell proliferation and viability can be assessed by utilizing methods that quantify the metabolic conversion of a natural or synthetic substrate (42). Other tests have been developed to interrogate the migratory and invasive capacities of cells, such as scratch-wound and zone exclusion assays (43); an empty “zone” is created into which motile metastatic cells migrate. The addition of an extracellular matrix (ECM) layer, such as Matrigel or collagen, mimics a basement membrane layer for cells to “invade” through, as occurs with transwell and Boyden chamber assays (43). These assays can be exploited further by incorporating a chemoattractant factor or chemotaxis. The concentration gradient of small molecules or chemokines can be lightly regulated as with modified transwell or capillary assays, or tightly controlled temporally and spatially with microfluidics (44, 45). Several studies have shown metastatic cells will follow a gradient or chemokine trail to “home” to their secondary destination (46, 47). For instance, the release of CXCL12 and CXCL16 by metastatic brain stroma has been shown to attract breast cancer cells (48). Likewise, CXCR4, CXCR7 and CXCL12 expression has been implicated in the homing of cancer cells to the brain (49, 50). Other cytokines can assist other stages of metastasis, such as aiding the transmigration of metastatic cells across the BBB (51), promoting vascular remodeling (52), and remodeling the metastatic cell cytoskeleton to enhance mobility (53). Time lapse microscopy allows for the observation of the morphological minutiae in cell motility, from determination of cell front velocity and zone closure speed, to the morphological changes a metastatic cell attains to identifying the type of motility (46, 54).

Table 1:

Major Assays for Metastasis

Select in vitro models
Model Description Advantages Limitations Reference
Invasion/Migration Assays Uses chambers with a wide range of chemical and properties to assess motility of cancers cell lines across physical distances Low costs for high cell throughputs; allows for comparisons between cancer cells for their metastatic potential under a variety of conditions Cells grow in 2D; cell migration in a petri dish may not reflect how metastasis occurs in living systems Katt et al.(32)
Tumorspheres Growing cancer cell lines in suspension or low binding plates to allow outgrowth in three dimensions Since cells grow in 3D, this model more accurately represents how both mechanical and chemical factors can affect tumor formation; effective for screening anti-metastasis drugs Many cell lines may not form tumorsphere structures, limiting this model’s use to only small subsets of cancer populations Lee et al. (33)
Organoids Three-dimensional cell culture technique: organoids are grown from embryonic or stem cells and resemble many of the phenotypes found in tissue of origin intercellular communication networks and biological signaling are similar to that of original organ, allowing for more accurate drug screen assays Random and uncontrolled growth of organoid cancer cells is common; does not accurately reflect cell communication from cells outside the original organ Drost et al.(34); Hynds et al. (35); Kim et al. (36); Klein et al.(37)
Select in vitro models
Model Description Advantages Limitations Reference
Syngeneic Model Engraft cancer cells that derive from the same species as the host animal Animal retains fully competent immune system; effective for testing new immunotherapies Does not always reflect human immune system dynamics, since animal immune system is fundamentally different Saito et al. (38)
Humanized Mouse Model Engraft human immune system issue into immune-deficient mice Fast experimental kinetics; reflects human-like immune environment in animals Humanized mice have high mortality rates than wild type mice. Grafting efficiency can also be variable from mouse to mouse. Yin et al.(39)
Genetically Engineered Mouse Model Knockdown/upregulation of select genes in a model mouse Allows for isolation and subsequent study of specific genes/oncogenic pathways Expensive; Genetically engineered mouse lines also take a long time to establish. Walrath et al.(40)
Xenografts Engraft human tumor tissue into humanized or immunodeficient mice Reflects the tumor microenvironment of the original cancer sample accurately. Many tumor types will not successfully engraft or metastasize after implantation Saito et al. (38)

These in vitro assays are largely two-dimensional (2D), and so lack significant factors such as the microenvironmental influence and cell-cell interactions. These deficiencies limit the translational potential of these assays. Significant efforts have been expended on developing three-dimensional (3D) assays to retain these essential interactions, such as incorporation of stromal cells (immune cells, endothelial cells, fibroblasts, etc.)(55) and enhanced matrices and scaffolds that allow adhesion (56).

Tumorspheres are spherical structures generated by cancer stem cells (CSCs). Studies have shown that this particular subpopulation within the bulk tumor possesses self-renewal properties leading to tumor recurrence, differentiation, metastasis and drug resistance (33, 57). Methods involve culturing cell lines and allowing them to aggregate on low-binding plates or hanging in drops of serum-free media and a cocktail of growth factors. The screening of potential anti-metastatic agents is more translatable to in vivo anti-tumorigenic activity when conducted with tumorspheres as compared to 2D monolayer cultures (33). Many small molecule inhibitors of components of signaling pathways vital to CSCs have entered clinical trials, such as GDC-0084 and Regorafenib, and others have gained FDA approval, such as RO4929097 (58), sonidegib (59), and glasdegib (60, 61).

Organoids, which are derived from tumor biopsies or resections, are characterized by their ability to self-organize into hierarchies that reflect their derivative tissues (34). Organoids have greater similarities to patient samples as compared to cells grown under 2D culturing conditions (62). A recently described procedure utilizing human cancer cells and human embryonic stem cell-derived cerebral organoids can successfully reproduce metastatic cancer processes and serve as a useful tool for drug screening, with lung-specific X protein (LUNX) found to play an important role in cell proliferation and migration or invasion (63).

Organotypic assays evaluate tumor cell invasion into brain tissue and offer an alternative to in vivo models. A few variations of these models have been developed. Typically, a mouse brain is sliced to a specified thickness, cultured at an air/liquid interface, and tumor cells or spheroids either plated right beside, or implanted directly into, the slice. The objective of this assay is to track cell infiltration into the neural tissue and thereby provide a better evaluation of metastatic cell invasion as it encounters the extracellular matrix (64). Organotypic assays have proven superior to 2D models as they take into account the unique ECM composition in the brain, maintaining their normal cytoarchitecture, complex cell relationships and biochemical and electrophysiological properties, while avoiding the costly, labor intensive and lengthy study periods associated with in vivo models (41, 65, 66).

The type of cell line selected for in vitro assays driven by the experimental endpoint. Commercial cell lines are readily available, with culture conditions known and that are characterized by relatively simple maintenance and rapid growth rates. Commercial cell lines are also often easy use for genetic manipulation, such as for loss-of or gain-of function studies and to allow clonal selection and expansion to select for transduced populations (67). However, as these cell lines have been developed over years or decades in culture conditions lacking the original microenvironmental cues and factors, they represent only a small, homogenous subpopulation of the original patient samples, thereby lacking the original tumor heterogeneity and displaying different genetic and epigenetic characteristics (68). Additionally, while the use of these cell lines should theoretically enable the generation of results that are comparable across laboratories, this has not been found to be the case, with many reports of inconsistencies reported for drug sensitivity (69, 70). Primary patient-derived cell lines are low passage lines developed from patient tumor samples. These lines more accurately reflect the molecular characteristics of the original patient tumor than do commercial cell lines. Because these model systems display better accuracy at predicting the success rates of potential therapeutics, they may be useful in making clinical decisions and in improving the field of personalized medicine (67, 71). However, use of patient-derived cell lines is hindered by the difficulties associated with obtaining enough samples or cells for experiments and the long latency periods for tumor development in vivo.

These in vitro models are valuable tools for elucidating functional and biological metastatic properties that may not be detectible with in vivo models. These assays are rapid, easy to perform, relatively inexpensive and lend themselves to high throughput drug screening. Additionally, by focusing on single metastatic stages these assays allow for an assessment of the influence individual molecules and genes by making it possible to genetically manipulate specific steps of tumor progression and metastasis (67). However, given the limitations of in vitro assays, the resultant data may not accurately reflect what occurs in vivo.

In vivo models of metastasis

The value of an in vivo model in identifying novel therapeutic candidates is the ability of the model to replicate the clinical progression of the disease in question (Table 1). Rodents (typically mice) have proven to be especially valuable for developing models that advance our understanding of the pathophysiology and etiology of cancer (72) and for the evaluation of novel candidate therapies (73). A number of patient-derived xenograft (PDX) models have been generated for studying brain metastasis. With these in vivo models, PDXs can retain better tumor heterogeneity, biology and microenvironmental conditions as compared to in vitro models (74). Use of these in vivo models has led to the proposition of several key genes and pathways that are essential to metastatic progression(75) as well as the identification of novel drug candidates for the treatment of this condition (67). Several varieties of spontaneous, induced, and experimental brain metastasis models have been established with commercial and patient-derived cell lines. These include tissues of both human and mouse origins, and involve cancers induced by genetic engineering, chemical or UV exposure. The route of cell inoculation can also determine the location of metastatic formation (76).

1. Syngeneic

Several established preclinical models have led to the discovery of key genes and pathways that are essential to metastatic progression. The syngeneic mouse model is one of the oldest models of brain metastasis and involves injecting allografts from immortalized murine cancer cells directly into mice (76). Both the cell donor mouse and host mouse typically come from the same inbred strains. Since the species of the cell line and the host animal match, syngeneic models allow the mouse to retain a fully competent immune system (77). This feature ensures that syngeneic models are particularly well suited for studying the efficacy of immunotherapies as well as interactions between cancer and immune cells (77). Commonly used murine cell lines for assessing brain derived cancers include GL261, CT-2A, and SMA-560 for glioma syngeneic models, all of which have been used for studying response to immunotherapy(7880).

With spontaneous syngeneic mouse models, the engrafted cell lines are derived directly from cancers that occur naturally in mice (81). The primary advantage of this model is quick progression of tumor metastasis in the host immunocompetent mouse. With a standard genetic mouse model of cancer progression, a brain metastasis assay could take months to complete (81). Spontaneous syngeneic mouse models for studying brain metastasis typically employ three neurotrophic cancer subtypes: melanoma, lung, and breast (82). Examples of commonly used spontaneous cancer cell lines used to study brain metastasis include 4T1 mammary gland tissue (83), WM239 melanoma (84), and 113/6–4L melanoma (84). The 4TI spontaneous model was used, for example, to establish ID2 as a promoter of breast cancer brain metastasis, a promising therapeutic target (83). Similarly, the WM239 melanoma model allowed for the identification of endothelin receptor B (ENDRB) as a potential target for treating melanoma brain metastasis (85). In addition to identifying genetic markers, spontaneous syngeneic models are used to discover novel drugs for treating brain metastasis patients. The tyrosine-kinase inhibitor Neratinib, used for treating patients with Her2 amplified breast cancer, inhibited brain metastasis when tested in spontaneous syngeneic experiments (86).

Induced syngeneic mouse models differ from spontaneous models in that the engrafted cancer cell lines come from healthy animals exposed to carcinogens (76). Chemical and UV exposure are two common techniques used to induce cancer in the donor animals (76). Chemical induction of lung cancer can be performed by injection of urethane and nicotine-derived nitrosamine ketone (NNK), while breast and skin cancers are induced by 7,12-Dimethylbenz(a)anthracene (DMBA) oral gavage and topical application, respectively (87). Ultraviolet exposure remains the most effective method for inducing murine melanoma, with the K-1735 cell line serving as a model system for studying brain metastasis (88). Syngeneic models with K-1735 cell lines were utilized to highlight the role of cytokines such as TGF-beta2 in facilitating melanoma metastasis (89), which ultimately led for TGF-beta2 inhibitors like Fresolimumab and Cemiplimab (90). Chemically induced syngeneic models have a number of advantages for preclinical testing, from their reproducibility to strong similarities with the human condition (91). Typically, syngeneic mouse model experiments for studying brain metastases typically use spontaneous or UV-induced cancer cell lines.

2. Humanized

Humanized mouse models are generated by engrafting human-derived immune system tissue and immune cells into immunodeficient mice. The model follows the fast kinetics of syngeneic mouse experiments while also reflecting a human-like immune environment in the mouse (92). The mouse and human immune system are fundamentally different, with mice having lower activation of the innate system and more naïve lymphocytes (93). As many immune system drugs are species-specific, humanized mouse models present clear advantages for pre-clinical testing by allowing for the laboratory testing of human immune system drugs (92). Because immunotherapy is a powerful treatment option for managing some brain metastases, humanized mouse models provide an excellent platform for testing new CNS metastasis drugs (94).

The Hu-PBL-NSG mouse model is a popular humanized model for evaluating immuno-oncology drugs. It is created by engrafting peripheral blood mononuclear cells (PBMCs) into immunodeficient NSG strain mice (95). Humanized mouse models were used to test the efficacy of anti-PD-1 immune checkpoint inhibitor drugs such as pembrolizumab in primary tumors of melanoma and lung cancer (95, 96). These immune checkpoint inhibitors are now employed for treating CNS metastases. In a study of patients with melanoma and lung CNS metastases, pembrolizumab showed promising activity in the brain (97, 98). Other immunotherapies such as ipilimumab and nivolumab were also initially developed using pre-clinical mouse models and are now used to treat melanoma brain CNS metastases (99, 100). These new immunotherapy drugs highlight the importance of humanized mouse models in shaping clinical practice.

3. GEMM

Genetically engineered mouse models (GEMMs) are divided into two categories: transgenic GEMM, where exogenous oncogenes are expressed through inoculation of a zygote or embryonic stem cells possessing the construct of the gene of interest, and targeted GEMM, incorporating homologous recombination in mouse embryonic stem cells (101, 102)). With GEMMs tumors develop de novo in a natural immune-competent environment, where the histopathological and molecular features of the resulting tumor closely resemble those found in humans and are even able to spontaneously metastasize (102). The first set of such “onco-mice” models provided unequivocal proof that oncogenic expression in normal cells could lead to tumor development (103). The subsequent advancement in gene-targeting technology permitted the study of tumor suppressor gene (TSG) knockout mice (104, 105). TSGs are divided into two groups: “gatekeepers” that regulate tumor cell growth through control of proliferation and apoptosis, and “caretakers” that regulate cellular processes responsible for genetic alterations (106). These models, one of the most frequently utilized models in cancer research, have been developed for several important TSGs, including Rb (107), p53 (108, 109), Ink4a/Arf (110, 111), Brca1/2 (112, 113). The benefit to GEMMs is the ability for spontaneous metastatic development in an immunocompetent host, thereby mimicking the clinical condition, but also allowing for the study of micro-environmental and immunology components of the metastatic process (114). As syngeneic cell lines can also be applied to GEMMs, these models are of particular interest to those involved in brain metastasis research in relation to the immune system involvement. They also form brain metastasis more quickly than in PDX models (82, 115118).

4. Xenograft

The PDX models of brain metastasis are derived by transplanting fresh human tumor tissue or early passage patient-derived cells into immunodeficient or humanized mice; the lack of an immune system or immune system of the same species promotes higher tumor engraftment frequency. The benefit to PDX models is better retention of the genomic, histopathological and phenotypic heterogeneity of the original sample, thereby possibly improving the screening of potential therapies as well as their value in personalized medicine (119, 120). Currently, PDX models are utilized for large scale and high throughput drug screening studies to predict responses to clinical therapeutic candidates (121).

Routes of injection

The location of cell inoculation is key to the type of tumor that will develop and how closely the established model recapitulates the clinical condition. Tumor implantation in model locations other than the brain or primary organ are termed heterotopic or ectopic. Typically, the route of inoculation is intravenous (IV), where tumor cell dissemination will vary due to circulation. Delivery of cells through the tail vein will often result in metastases developing predominantly in the lung and subsequently to the CNS. Administration by this route has a very high survival rate and is easy to execute (122, 123).

To avoid pulmonary localization of cells, IV inoculation through the left heart ventricle, termed intracardiac, permits systemic distribution of cells throughout the body. Intracardiac models are useful in determining the homing propensity of tumor cells, where the seed vs. soil theory suggests that cells have a predisposition for certain secondary environments despite circulation (124). While not as robust as tail vein models, intracardiac models still have a high survival rate and the procedure is relatively simple to perform. To minimize the spread of tumor cells to tissues other than the brain, intracarotid artery inoculation is performed. This model can be time-intensive, requiring skills in microsurgery for the required ligation of the artery, and has a high intraprocedural mortality rate (76, 125).

For some IV studies, cells that have managed to metastasize to the brain are gathered and re-inoculated, creating cell lines that are “brain-trained”. This method ensures that cells home specifically to the brain despite the IV route used and can increase the frequency of metastatic formation in the brain with each cycle of inoculation (126, 127). Although the molecular mechanism for “brain-training” is unknown, these cells are likely more suited to undergo the same physical and chemical changes to cross through the blood-brain barrier again(128).

The model is termed orthotopic when cells are implanted into the originating tissue environment. In the case of brain metastasis, direct implantation of cells into the brain would more accurately be termed local growth models, whereas models that involve implanting cells into the primary organ of origin (lung, breast, skin, etc) more accurately reflect the metastatic cascade as metastases arise spontaneously (76). Experimental xenograft models have been developed for brain metastases derived from engrafting human lung (129, 130), breast (131, 132), and melanoma cells into mice (133, 134). However, local growth models of direct implantation into the brain are the most common, as generating tumor growth in the primary organ and subsequent brain metastasis is difficult to achieve. Direct implantation is typically accomplished utilizing stereotactic equipment to ensure accuracy of cell delivery into the neural tissue rather than the ventricles (122). Other techniques involve the use of a subarachnoid catheter placed into the cisterna magna along the spinal cord (135). Other techniques utilize a subarachnoid catheter placed into the cisterna magna along the spinal cord (117) or freehand delivery into landmarked areas of the right frontal lobe (136).

Subcutaneous inoculation, where cells are injected into the flank of a mouse, is a traditional route that is highly used in metastatic models because of the ease of the administration procedure. However, while this model is typically used to assess tumor engraftment, size, and therapy screening, it is only ideal in development of metastatic models from primary melanomas which originate in the skin. For any other model system, they do not reflect the complexity of the metastatic cascade or the multistage process of oncogenesis, and very frequently the cells injected do not originate from melanomas (137).

In vivo models for CNS drug discovery

With the currently limited understanding of the mechanisms and molecules involved in brain metastasis development, in vivo models are a vital tool in the discovery of new therapies for treating brain metastaseis. Identification of drug targets relevant to brain metastasis is a first step in developing effective therapies. While in vitro methods are more useful for the identification of targets and large-scale drug screening, in vivo methods are better suited for fine tuning target validation and compound efficacy (138). A

The BBB is a significant hindrance to developing clinically effective therapies for CNS disorders as the role of the BBB is to prevent access of toxins and other molecules, such as chemotherapeutics, into the CNS. This limits the number of therapies that can penetrate the BBB. Moreover, many of those that can have minimal effects on extending survival (139). In vivo models are critical in identifying effective and unique delivery systems to assist passage of chemotherapeutics across the BBB. Coating drugs with various nanoparticles (metallic, organic and biological) improves survival in mice with brain metastases by facilitating transfer into brain (140). Included in this group are nanoparticles with doxorubicin and lapatinib (141) or camptothecin/Herceptin (142), liposomes with simvastatin/gefitinib (143) or irinotecan (144), and dendrosomes with solanine (145) or curcumin (146). Gadolinium nanoparticles and radiotherapy are in Phase I clinical trials for patients with multiple brain metastases (Clinicaltrials.gov;NCT02820454). Another method involves disruption of the BBB itself with MRI-guided ultrasound disruption. This approach has been shown to improve delivery of antibodies such as trastuzumab (147), pertuzumab (148) and natural killer (NK-92) cells (149). Disruption of the BBB through the application of hypotonic solutions such as mannitol (150) are currently in clinical trial as a combined administration with methotrexate and carboplatin with/without trastuzumab (Clinicaltrials.gov; NCT00397501). Other therapies that have reached clinical trials based on preclinical brain metastasis data are the dual EGFR and HER2 targeting inhibitor lapatinib (151), ALTTO trial (152), (Clinicaltrials.gov;NCT00470847), the histone deacytelase HDAC inhibitor Vorinostat (153), (Clinicaltrials.gov; NCT00820222), a VEGFR-2 TKI Cediranib (154), (Clinicaltrials.gov;NCT00937482), a low-molecular-weight epothilone Sagolipone (155), (Clinicaltrials.gov; NCT00496379), and a Paclitaxel derivative ANG1005 (156), (Clinicaltrials.gov; NCT01679743)(157) (138).

Drug screening pitfalls

The success of in vivo models depends on their predictive value for the clinical setting. The most common reason for a compound to fail to demonstrate clinical efficacy is the lack of an association between the modulating activity of the specific target(s) and the disease state. Such a failure is essentially due to a lack of proper or accurate validation in the preclinical models or unclear relationship of the preclinical model to the condition for which the drug is tested (158). Approximately 95% of agents that pass preclinical validation fail in phase 1 of clinical development (159). As such, despite the wealth of knowledge and clinical advances achieved with in vivo modelling, there remain significant limitations to this approach in drug discovery. Most importantly, no preclinical model is known to mimic precisely the complete clinical presentation of brain metastases. In the screening of potential treatments, therapies are often administered very soon after tumor inoculation, which does not replicate when patients are treated with advanced metastatic disease of a size detectable by imaging (6, 160).

In contrast to in vitro assays, in vivo model experiments are much more difficult to perform on a large scale. The initial establishment of preclinical models is can also be time consuming and costly. Because the target transgenes are expressed in all cells of the specific tissue, TSG knockout mice often fail to mimic sporadically established cancers where the accumulation of genetic events in a single cell gives rise to a tumor in an otherwise normal organ. In syngeneic and GEMM models, the genetics of the model (promoter and oncogenes) are not truly representative of human disease and their specific regualtion and many chemically induced cell lines such as JB/MS melanoma do not spread to the brain readily and lose tumorigenicity rapidly. These models also require extensive and timely breeding programs (114).

The PDX models often involve long latency periods from tumor cell inoculation to metastatic initiation, or display inconsistent or no metastatic dissemination that results in a lack of robustness or reproducibility between cell lines. There are also concerns about genetic drift in PDX models generated with late passages of cell lines. However, several studies have shown no major genetic variation up to at least passage 10 (161, 162). Immunocompromised mice are necessary for tumor cell engraftment in PDX models, losing completely any influence the immune system would have on tumor growth, whereas syngeneic models replace the original human stroma and immune cells with mouse, thereby losing key similarities to human disease progression.

The route of inoculation plays a significant role in how accurately the model can represent and recapitulate the clinical condition. In the case of brain metastasis, local growth orthotopic models can replicate the later stages of metastasis of tumor engraftment in the brain and macro-metastatic growth, although the initial stages in the disease process are bypassed. Orthotopic models that involve implantation of the tumor cells into the primary tissue of origin (i.e., other than the brain) can facilitate spontaneous metastases formation. However, metastases are not always to the brain and there may be an extended lag in time from primary inoculation to metastases formation. These challenges are also common with intracardiac injections, where metastases can develop throughout the body and not solely in brain. Heterotopic models only incorporate the mid to end stages of metastasis, skipping the initial stages of escape from the primary tumor and intravasation. It remains to be seen whether models where metastases are generated chemically, genetically, or through UV exposure, accurately reflect clinical metastatic progression (76). The validity and accuracy of results comes into question when inappropriate models are utilized for drug screens. To date, few if any models properly replicate the progression of metastaic disease in humans (76).

Conclusion

In vitro and in vivo assays have led to significant advancements in deciphering the intricacies of metastasis. Cell culture methods are essential for interrogating key morphological and functional aspects of metastatic biology, with preclinical evaluation of novel anti-metastatic strategies in animal models being an essential step towards the development of novel therapeutics. However, the translation of experimental findings to the clinical arena remains a major bottleneck in the development of therapies for brain metastases. Despite these drawbacks, both in vitro and in vivo models provide significant contributions to the understanding of pathology of brain metastasis and remain a vital tool for the screening of novel therapeutics.

ACKNOWLEDGEMENTS:

PKB receives funding from Damon Runyon Cancer Research Foundation, Ben and Catherine Ivy Foundation, Breast Cancer Research Foundation, MGH Research Scholar Program, and the NCI (1R01CA227156-01, 5R21CA220253-02 and 1R01CA244975-01).

Footnotes

CONFLICT OF INTEREST STATEMENT:

Unrelated to this work, PKB has consulted for Tesaro, Angiochem, Genentech-Roche, ElevateBio, Eli Lilly, SK Life Sciences, Pfizer, Voyager Therapeutics and Dantari, received institutional research funding (to MGH) from Merck, Mirati, Eli Lilly, BMS and Pfizer and has received honoraria from Merck, Pfizer and Genentech-Roche.

References

  • 1.Nathoo N, Chahlavi A, Barnett GH, Toms SA. Pathobiology of brain metastases. Journal of clinical pathology. 2005;58(3):237–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Nayak L, Lee EQ, Wen PY. Epidemiology of brain metastases. Curr Oncol Rep. 2012;14(1):48–54. [DOI] [PubMed] [Google Scholar]
  • 3.Patchell RA. The management of brain metastases. Cancer Treat Rev. 2003;29(6):533–40. [DOI] [PubMed] [Google Scholar]
  • 4.Preusser M, Berghoff AS, Schadendorf D, Lin NU, Stupp R. Brain metastasis: opportunity for drug development? Curr Opin Neurol. 2012;25(6):786–94. [DOI] [PubMed] [Google Scholar]
  • 5.Barnholtz-Sloan JS, Yu C, Sloan AE, Vengoechea J, Wang M, Dignam JJ, et al. A nomogram for individualized estimation of survival among patients with brain metastasis. Neuro Oncol. 2012;14(7):910–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Palmieri D, Chambers AF, Felding-Habermann B, Huang S, Steeg PS. The biology of metastasis to a sanctuary site. Clin Cancer Res. 2007;13(6):1656–62. [DOI] [PubMed] [Google Scholar]
  • 7.Steeg PS, Camphausen KA, Smith QR. Brain metastases as preventive and therapeutic targets. Nat Rev Cancer. 2011;11(5):352–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Valiente M, Ahluwalia MS, Boire A, Brastianos PK, Goldberg SB, Lee EQ, et al. The Evolving Landscape of Brain Metastasis. Trends Cancer. 2018;4(3):176–96. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Tawbi HA, Forsyth PA, Algazi A, Hamid O, Hodi FS, Moschos SJ, et al. Combined Nivolumab and Ipilimumab in Melanoma Metastatic to the Brain. N Engl J Med. 2018;379(8):722–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Long GV, Atkinson V, Lo S, Sandhu S, Guminski AD, Brown MP, et al. Combination nivolumab and ipilimumab or nivolumab alone in melanoma brain metastases: a multicentre randomised phase 2 study. Lancet Oncol. 2018;19(5):672–81. [DOI] [PubMed] [Google Scholar]
  • 11.Park S, Lee MH, Seong M, Kim ST, Kang JH, Cho BC, et al. A phase II, multicenter, two cohort study of 160 mg osimertinib in EGFR T790M-positive non-small-cell lung cancer patients with brain metastases or leptomeningeal disease who progressed on prior EGFR TKI therapy. Ann Oncol. 2020;31(10):1397–404. [DOI] [PubMed] [Google Scholar]
  • 12.Lin NU, Borges V, Anders C, Murthy RK, Paplomata E, Hamilton E, et al. Intracranial Efficacy and Survival With Tucatinib Plus Trastuzumab and Capecitabine for Previously Treated HER2-Positive Breast Cancer With Brain Metastases in the HER2CLIMB Trial. J Clin Oncol. 2020;38(23):2610–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Davies MA, Saiag P, Robert C, Grob JJ, Flaherty KT, Arance A, et al. Dabrafenib plus trametinib in patients with BRAF(V600)-mutant melanoma brain metastases (COMBI-MB): a multicentre, multicohort, open-label, phase 2 trial. Lancet Oncol. 2017;18(7):863–73. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Ni W, Chen W, Lu Y. Emerging findings into molecular mechanism of brain metastasis. Cancer Med. 2018;7(8):3820–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Chambers AF, Groom AC, MacDonald IC. Dissemination and growth of cancer cells in metastatic sites. Nat Rev Cancer. 2002;2(8):563–72. [DOI] [PubMed] [Google Scholar]
  • 16.Popper H Primary tumor and metastasis-sectioning the different steps of the metastatic cascade. Transl Lung Cancer Res. 2020;9(5):2277–300. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Eichler AF, Chung E, Kodack DP, Loeffler JS, Fukumura D, Jain RK. The biology of brain metastases-translation to new therapies. Nat Rev Clin Oncol. 2011;8(6):344–56. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Martin TA, Ye L, Sanders AJ, Lane J, Jiang WG. Cancer Invasion and Metastasis: Molecular and Cellular Perspective: Landes Bioscience; 2013.
  • 19.Kim YN, Koo KH, Sung JY, Yun UJ, Kim H. Anoikis resistance: an essential prerequisite for tumor metastasis. Int J Cell Biol. 2012;2012:306879. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Yang Y, Wang Y, Che X, Hou K, Wu J, Zheng C, et al. Integrin alpha5 promotes migration and invasion through the FAK/STAT3/AKT signaling pathway in icotinib-resistant non-small cell lung cancer cells. Oncol Lett. 2021;22(1):556. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Chiang SP, Cabrera RM, Segall JE. Tumor cell intravasation. Am J Physiol Cell Physiol. 2016;311(1):C1–C14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Liu Y, Cao X. Immunosuppressive cells in tumor immune escape and metastasis. J Mol Med (Berl). 2016;94(5):509–22. [DOI] [PubMed] [Google Scholar]
  • 23.Fares J, Fares MY, Khachfe HH, Salhab HA, Fares Y. Molecular principles of metastasis: a hallmark of cancer revisited. Signal Transduct Target Ther. 2020;5(1):28. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Liu Q, Zhang H, Jiang X, Qian C, Liu Z, Luo D. Factors involved in cancer metastasis: a better understanding to “seed and soil” hypothesis. Mol Cancer. 2017;16(1):176. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Daneman R, Prat A. The blood-brain barrier. Cold Spring Harb Perspect Biol. 2015;7(1):a020412. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Wilhelm I, Molnar J, Fazakas C, Hasko J, Krizbai IA. Role of the blood-brain barrier in the formation of brain metastases. Int J Mol Sci. 2013;14(1):1383–411. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Tiwary S, Morales JE, Kwiatkowski SC, Lang FF, Rao G, McCarty JH. Metastatic Brain Tumors Disrupt the Blood-Brain Barrier and Alter Lipid Metabolism by Inhibiting Expression of the Endothelial Cell Fatty Acid Transporter Mfsd2a. Sci Rep. 2018;8(1):8267. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Belykh E, Shaffer KV, Lin C, Byvaltsev VA, Preul MC, Chen L. Blood-Brain Barrier, Blood-Brain Tumor Barrier, and Fluorescence-Guided Neurosurgical Oncology: Delivering Optical Labels to Brain Tumors. Front Oncol. 2020;10:739. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Carbonell WS, Ansorge O, Sibson N, Muschel R. The vascular basement membrane as “soil” in brain metastasis. PLoS One. 2009;4(6):e5857. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Fortin D The blood-brain barrier: its influence in the treatment of brain tumors metastases. Curr Cancer Drug Targets. 2012;12(3):247–59. [DOI] [PubMed] [Google Scholar]
  • 31.Menezes ME, Das SK, Minn I, Emdad L, Wang XY, Sarkar D, et al. Detecting Tumor Metastases: The Road to Therapy Starts Here. Adv Cancer Res. 2016;132:1–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Katt ME, Placone AL, Wong AD, Xu ZS, Searson PC. In Vitro Tumor Models: Advantages, Disadvantages, Variables, and Selecting the Right Platform. Front Bioeng Biotechnol. 2016;4:12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Lee CH, Yu CC, ssssWang BY, Chang WW. Tumorsphere as an effective in vitro platform for screening anti-cancer stem cell drugs. Oncotarget. 2016;7(2):1215–26. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Drost J, Clevers H. Organoids in cancer research. Nat Rev Cancer. 2018;18(7):407–18. [DOI] [PubMed] [Google Scholar]
  • 35.Hynds RE, Giangreco A. Concise review: the relevance of human stem cell-derived organoid models for epithelial translational medicine. Stem Cells. 2013;31(3):417–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Kim J, Koo BK, Knoblich JA. Human organoids: model systems for human biology and medicine. Nat Rev Mol Cell Biol. 2020;21(10):571–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Klein E, Hau AC, Oudin A, Golebiewska A, Niclou SP. Glioblastoma Organoids: Pre-Clinical Applications and Challenges in the Context of Immunotherapy. Front Oncol. 2020;10:604121. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Saito R, Kobayashi T, Kashima S, Matsumoto K, Ogawa O. Faithful preclinical mouse models for better translation to bedside in the field of immuno-oncology. Int J Clin Oncol. 2020;25(5):831–41. [DOI] [PubMed] [Google Scholar]
  • 39.Yin L, Wang XJ, Chen DX, Liu XN, Wang XJ. Humanized mouse model: a review on preclinical applications for cancer immunotherapy. Am J Cancer Res. 2020;10(12):4568–84. [PMC free article] [PubMed] [Google Scholar]
  • 40.Walrath JC, Hawes JJ, Van Dyke T, Reilly KM. Genetically engineered mouse models in cancer research. Adv Cancer Res. 2010;106:113–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Grotzer MA, Neve A, Baumgartner M. Dissecting brain tumor growth and metastasis in vitro and ex vivo. Journal of Cancer Metastasis and Treatment. 2016;2:149–62. [Google Scholar]
  • 42.Romar GA, Kupper TS, Divito SJ. Research Techniques Made Simple: Techniques to Assess Cell Proliferation. J Invest Dermatol. 2016;136(1):e1–e7. [DOI] [PubMed] [Google Scholar]
  • 43.Kramer N, Walzl A, Unger C, Rosner M, Krupitza G, Hengstschlager M, et al. In vitro cell migration and invasion assays. Mutat Res. 2013;752(1):10–24. [DOI] [PubMed] [Google Scholar]
  • 44.Sontheimer-Phelps A, Hassell BA, Ingber DE. Modelling cancer in microfluidic human organs-on-chips. Nat Rev Cancer. 2019;19(2):65–81. [DOI] [PubMed] [Google Scholar]
  • 45.Mosadegh B, Lockett MR, Minn KT, Simon KA, Gilbert K, Hillier S, et al. A paper-based invasion assay: assessing chemotaxis of cancer cells in gradients of oxygen. Biomaterials. 2015;52:262–71. [DOI] [PubMed] [Google Scholar]
  • 46.Huth J, Buchholz M, Kraus JM, Schmucker M, von Wichert G, Krndija D, et al. Significantly improved precision of cell migration analysis in time-lapse video microscopy through use of a fully automated tracking system. BMC Cell Biol. 2010;11:24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Fares J, Cordero A, Kanojia D, Lesniak MS. The Network of Cytokines in Brain Metastases. Cancers (Basel). 2021;13(1). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Chung B, Esmaeili AA, Gopalakrishna-Pillai S, Murad JP, Andersen ES, Kumar Reddy N, et al. Human brain metastatic stroma attracts breast cancer cells via chemokines CXCL16 and CXCL12. NPJ Breast Cancer. 2017;3:6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Salmaggi A, Maderna E, Calatozzolo C, Gaviani P, Canazza A, Milanesi I, et al. CXCL12, CXCR4 and CXCR7 expression in brain metastases. Cancer Biol Ther. 2009;8(17):1608–14. [DOI] [PubMed] [Google Scholar]
  • 50.Sun X, Cheng G, Hao M, Zheng J, Zhou X, Zhang J, et al. CXCL12 / CXCR4 / CXCR7 chemokine axis and cancer progression. Cancer Metastasis Rev. 2010;29(4):709–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Rai S, Nejadhamzeeigilani Z, Gutowski NJ, Whatmore JL. Loss of the endothelial glycocalyx is associated with increased E-selectin mediated adhesion of lung tumour cells to the brain microvascular endothelium. J Exp Clin Cancer Res. 2015;34:105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Rodrigues G, Hoshino A, Kenific CM, Matei IR, Steiner L, Freitas D, et al. Tumour exosomal CEMIP protein promotes cancer cell colonization in brain metastasis. Nat Cell Biol. 2019;21(11):1403–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Zhang L, Liu J, Wang X, Li Z, Zhang X, Cao P, et al. Upregulation of cytoskeleton protein and extracellular matrix protein induced by stromal-derived nitric oxide promotes lung cancer invasion and metastasis. Curr Mol Med. 2014;14(6):762–71. [DOI] [PubMed] [Google Scholar]
  • 54.Jain P, Worthylake RA, Alahari SK. Quantitative analysis of random migration of cells using time-lapse video microscopy. J Vis Exp. 2012(63):e3585. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Infanger DW, Lynch ME, Fischbach C. Engineered culture models for studies of tumor-microenvironment interactions. Annu Rev Biomed Eng. 2013;15:29–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Kenney RM, Loeser A, Whitman NA, Lockett MR. Paper-based Transwell assays: an inexpensive alternative to study cellular invasion. Analyst. 2018;144(1):206–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Yelle N, Bakhshinyan D, Venugopal C, Singh SK. Introduction to Brain Tumor Stem Cells. Methods Mol Biol. 2019;1869:1–9. [DOI] [PubMed] [Google Scholar]
  • 58.Xu R, Shimizu F, Hovinga K, Beal K, Karimi S, Droms L, et al. Molecular and Clinical Effects of Notch Inhibition in Glioma Patients: A Phase 0/I Trial. Clin Cancer Res. 2016;22(19):4786–96. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Cazet AS, Hui MN, Elsworth BL, Wu SZ, Roden D, Chan CL, et al. Targeting stromal remodeling and cancer stem cell plasticity overcomes chemoresistance in triple negative breast cancer. Nat Commun. 2018;9(1):2897. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Du FY, Zhou QF, Sun WJ, Chen GL. Targeting cancer stem cells in drug discovery: Current state and future perspectives. World J Stem Cells. 2019;11(7):398–420. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Zhou C, Du J, Zhao L, Liu W, Zhao T, Liang H, et al. GLI1 reduces drug sensitivity by regulating cell cycle through PI3K/AKT/GSK3/CDK pathway in acute myeloid leukemia. Cell Death & Disease. 2021;12(3):231. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Duarte AA, Gogola E, Sachs N, Barazas M, Annunziato S, J RdR, et al. BRCA-deficient mouse mammary tumor organoids to study cancer-drug resistance. Nat Methods. 2018;15(2):134–40. [DOI] [PubMed] [Google Scholar]
  • 63.Choe MS, Kim JS, Yeo HC, Bae CM, Han HJ, Baek K, et al. A simple metastatic brain cancer model using human embryonic stem cell-derived cerebral organoids. FASEB J. 2020;34(12):16464–75. [DOI] [PubMed] [Google Scholar]
  • 64.Jung S, Kim HW, Lee JH, Kang SS, Rhu HH, Jeong YI, et al. Brain tumor invasion model system using organotypic brain-slice culture as an alternative to in vivo model. J Cancer Res Clin Oncol. 2002;128(9):469–76. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Shamir ER, Ewald AJ. Three-dimensional organotypic culture: experimental models of mammalian biology and disease. Nat Rev Mol Cell Biol. 2014;15(10):647–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Humpel C Organotypic brain slice cultures: A review. Neuroscience. 2015;305:86–98. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Huo KG, D’Arcangelo E, Tsao MS. Patient-derived cell line, xenograft and organoid models in lung cancer therapy. Transl Lung Cancer Res. 2020;9(5):2214–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Wilding JL, Bodmer WF. Cancer cell lines for drug discovery and development. Cancer Res. 2014;74(9):2377–84. [DOI] [PubMed] [Google Scholar]
  • 69.Haibe-Kains B, El-Hachem N, Birkbak NJ, Jin AC, Beck AH, Aerts HJ, et al. Inconsistency in large pharmacogenomic studies. Nature. 2013;504(7480):389–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Cancer Cell Line Encyclopedia C, Genomics of Drug Sensitivity in Cancer C. Pharmacogenomic agreement between two cancer cell line data sets. Nature. 2015;528(7580):84–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Kodack DP, Farago AF, Dastur A, Held MA, Dardaei L, Friboulet L, et al. Primary Patient-Derived Cancer Cells and Their Potential for Personalized Cancer Patient Care. Cell Rep. 2017;21(11):3298–309. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. 2011;144(5):646–74. [DOI] [PubMed] [Google Scholar]
  • 73.Day CP, Merlino G, Van Dyke T. Preclinical mouse cancer models: a maze of opportunities and challenges. Cell. 2015;163(1):39–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Cassidy JW, Caldas C, Bruna A. Maintaining Tumor Heterogeneity in Patient-Derived Tumor Xenografts. Cancer Res. 2015;75(15):2963–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Shih DJH, Nayyar N, Bihun I, Dagogo-Jack I, Gill CM, Aquilanti E, et al. Genomic characterization of human brain metastases identifies drivers of metastatic lung adenocarcinoma. Nat Genet. 2020;52(4):371–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Daphu I, Sundstrom T, Horn S, Huszthy PC, Niclou SP, Sakariassen PO, et al. In vivo animal models for studying brain metastasis: value and limitations. Clin Exp Metastasis. 2013;30(5):695–710. [DOI] [PubMed] [Google Scholar]
  • 77.Zhong W, Myers JS, Wang F, Wang K, Lucas J, Rosfjord E, et al. Comparison of the molecular and cellular phenotypes of common mouse syngeneic models with human tumors. BMC Genomics. 2020;21(1):2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Binello E, Qadeer ZA, Kothari HP, Emdad L, Germano IM. Stemness of the CT-2A Immunocompetent Mouse Brain Tumor Model: Characterization In Vitro. J Cancer. 2012;3:166–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Oh T, Fakurnejad S, Sayegh ET, Clark AJ, Ivan ME, Sun MZ, et al. Immunocompetent murine models for the study of glioblastoma immunotherapy. J Transl Med. 2014;12:107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Sanchez VE, Lynes JP, Walbridge S, Wang X, Edwards NA, Nwankwo AK, et al. GL261 luciferase-expressing cells elicit an anti-tumor immune response: an evaluation of murine glioma models. Sci Rep. 2020;10(1):11003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Paschall AV, Liu K. An Orthotopic Mouse Model of Spontaneous Breast Cancer Metastasis. J Vis Exp. 2016(114). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Valiente M, Van Swearingen AED, Anders CK, Bairoch A, Boire A, Bos PD, et al. Brain Metastasis Cell Lines Panel: A Public Resource of Organotropic Cell Lines. Cancer Res. 2020;80(20):4314–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Kijewska M, Viski C, Turrell F, Fitzpatrick A, van Weverwijk A, Gao Q, et al. Using an in-vivo syngeneic spontaneous metastasis model identifies ID2 as a promoter of breast cancer colonisation in the brain. Breast Cancer Res. 2019;21(1):4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Francia G, Cruz-Munoz W, Man S, Xu P, Kerbel RS. Mouse models of advanced spontaneous metastasis for experimental therapeutics. Nat Rev Cancer. 2011;11(2):135–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Cruz-Munoz W, Jaramillo ML, Man S, Xu P, Banville M, Collins C, et al. Roles for endothelin receptor B and BCL2A1 in spontaneous CNS metastasis of melanoma. Cancer Res. 2012;72(19):4909–19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Nagpal A, Redvers RP, Ling X, Ayton S, Fuentes M, Tavancheh E, et al. Neoadjuvant neratinib promotes ferroptosis and inhibits brain metastasis in a novel syngeneic model of spontaneous HER2(+ve) breast cancer metastasis. Breast Cancer Res. 2019;21(1):94. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Kemp CJ. Animal Models of Chemical Carcinogenesis: Driving Breakthroughs in Cancer Research for 100 Years. Cold Spring Harb Protoc. 2015;2015(10):865–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Radinsky R, Beltran PJ, Tsan R, Zhang R, Cone RD, Fidler IJ. Transcriptional induction of the melanocyte-stimulating hormone receptor in brain metastases of murine K-1735 melanoma. Cancer Res. 1995;55(1):141–8. [PubMed] [Google Scholar]
  • 89.Zhang C, Zhang F, Tsan R, Fidler IJ. Transforming growth factor-beta2 is a molecular determinant for site-specific melanoma metastasis in the brain. Cancer Res. 2009;69(3):828–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Ciardiello D, Elez E, Tabernero J, Seoane J. Clinical development of therapies targeting TGFbeta: current knowledge and future perspectives. Ann Oncol. 2020;31(10):1336–49. [DOI] [PubMed] [Google Scholar]
  • 91.Pearson HB, Pouliot N. Modeling Metastasis In Vivo. Jandial R, editor: Landes Biosciences; 2013. [Google Scholar]
  • 92.Walsh NC, Kenney LL, Jangalwe S, Aryee KE, Greiner DL, Brehm MA, et al. Humanized Mouse Models of Clinical Disease. Annu Rev Pathol. 2017;12:187–215. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Tao L, Reese TA. Making Mouse Models That Reflect Human Immune Responses. Trends Immunol. 2017;38(3):181–93. [DOI] [PubMed] [Google Scholar]
  • 94.Nieblas-Bedolla E, Nayyar N, Singh M, Sullivan RJ, Brastianos PK. Emerging Immunotherapies in the Treatment of Brain Metastases. Oncologist. 2021;26(3):231–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Pyo KH, Kim JH, Lee JM, Kim SE, Cho JS, Lim SM, et al. Promising preclinical platform for evaluation of immuno-oncology drugs using Hu-PBL-NSG lung cancer models. Lung Cancer. 2019;127:112–21. [DOI] [PubMed] [Google Scholar]
  • 96.Kametani Y, Ohno Y, Ohshima S, Tsuda B, Yasuda A, Seki T, et al. Humanized Mice as an Effective Evaluation System for Peptide Vaccines and Immune Checkpoint Inhibitors. Int J Mol Sci. 2019;20(24). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Goldberg SB, Schalper KA, Gettinger SN, Mahajan A, Herbst RS, Chiang AC, et al. Pembrolizumab for management of patients with NSCLC and brain metastases: long-term results and biomarker analysis from a non-randomised, open-label, phase 2 trial. Lancet Oncol. 2020;21(5):655–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Wang S, Hu C, Xie F, Liu Y. Use of Programmed Death Receptor-1 and/or Programmed Death Ligand 1 Inhibitors for the Treatment of Brain Metastasis of Lung Cancer. Onco Targets Ther. 2020;13:667–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Amaral T, Kiecker F, Schaefer S, Stege H, Kaehler K, Terheyden P, et al. Combined immunotherapy with nivolumab and ipilimumab with and without local therapy in patients with melanoma brain metastasis: a DeCOG* study in 380 patients. J Immunother Cancer. 2020;8(1). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Selby MJ, Engelhardt JJ, Johnston RJ, Lu LS, Han M, Thudium K, et al. Preclinical Development of Ipilimumab and Nivolumab Combination Immunotherapy: Mouse Tumor Models, In Vitro Functional Studies, and Cynomolgus Macaque Toxicology. PLoS One. 2016;11(9):e0161779. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Khaled WT, Liu P. Cancer mouse models: past, present and future. Semin Cell Dev Biol. 2014;27:54–60. [DOI] [PubMed] [Google Scholar]
  • 102.Kersten K, de Visser KE, van Miltenburg MH, Jonkers J. Genetically engineered mouse models in oncology research and cancer medicine. EMBO Mol Med. 2017;9(2):137–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Hanahan D, Wagner EF, Palmiter RD. The origins of oncomice: a history of the first transgenic mice genetically engineered to develop cancer. Genes Dev. 2007;21(18):2258–70. [DOI] [PubMed] [Google Scholar]
  • 104.Finlay CA. p53 loss of function: implications for the processes of immortalization and tumorigenesis. Bioessays. 1992;14(8):557–60. [DOI] [PubMed] [Google Scholar]
  • 105.Jonkers J, Berns A. Conditional mouse models of sporadic cancer. Nat Rev Cancer. 2002;2(4):251–65. [DOI] [PubMed] [Google Scholar]
  • 106.Taneja P, Zhu S, Maglic D, Fry EA, Kendig RD, Inoue K. Transgenic and knockout mice models to reveal the functions of tumor suppressor genes. Clin Med Insights Oncol. 2011;5:235–57. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Bignon YJ, Chen Y, Chang CY, Riley DJ, Windle JJ, Mellon PL, et al. Expression of a retinoblastoma transgene results in dwarf mice. Genes Dev. 1993;7(9):1654–62. [DOI] [PubMed] [Google Scholar]
  • 108.Frese KK, Tuveson DA. Maximizing mouse cancer models. Nat Rev Cancer. 2007;7(9):645–58. [DOI] [PubMed] [Google Scholar]
  • 109.Donehower LA, Harvey M, Slagle BL, McArthur MJ, Montgomery CA Jr., Butel JS, et al. Mice deficient for p53 are developmentally normal but susceptible to spontaneous tumours. Nature. 1992;356(6366):215–21. [DOI] [PubMed] [Google Scholar]
  • 110.Serrano M, Lee H, Chin L, Cordon-Cardo C, Beach D, DePinho RA. Role of the INK4a locus in tumor suppression and cell mortality. Cell. 1996;85(1):27–37. [DOI] [PubMed] [Google Scholar]
  • 111.Yang C, Ionescu-Tiba V, Burns K, Gadd M, Zukerberg L, Louis DN, et al. The role of the cyclin D1-dependent kinases in ErbB2-mediated breast cancer. Am J Pathol. 2004;164(3):1031–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Hoshino A, Yee CJ, Campbell M, Woltjer RL, Townsend RL, van der Meer R, et al. Effects of BRCA1 transgene expression on murine mammary gland development and mutagen-induced mammary neoplasia. Int J Biol Sci. 2007;3(5):281–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Hakem R, de la Pompa JL, Mak TW. Developmental studies of Brca1 and Brca2 knock-out mice. J Mammary Gland Biol Neoplasia. 1998;3(4):431–45. [DOI] [PubMed] [Google Scholar]
  • 114.Gomez-Cuadrado L, Tracey N, Ma R, Qian B, Brunton VG. Mouse models of metastasis: progress and prospects. Dis Model Mech. 2017;10(9):1061–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Taggart D, Andreou T, Scott KJ, Williams J, Rippaus N, Brownlie RJ, et al. Anti-PD-1/anti-CTLA-4 efficacy in melanoma brain metastases depends on extracranial disease and augmentation of CD8(+) T cell trafficking. Proc Natl Acad Sci U S A. 2018;115(7):E1540–E9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Limberis MP, Bell CL, Wilson JM. Identification of the murine firefly luciferase-specific CD8 T-cell epitopes. Gene Ther. 2009;16(3):441–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Hall DG, Stoica G. Characterization of brain and bone-metastasizing clones selected from an ethylnitrosourea-induced rat mammary carcinoma. Clin Exp Metastasis. 1994;12(4):283–95. [DOI] [PubMed] [Google Scholar]
  • 118.Kircher DA, Trombetti KA, Silvis MR, Parkman GL, Fischer GM, Angel SN, et al. AKT1(E17K) Activates Focal Adhesion Kinase and Promotes Melanoma Brain Metastasis. Mol Cancer Res. 2019;17(9):1787–800. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Siolas D, Hannon GJ. Patient-derived tumor xenografts: transforming clinical samples into mouse models. Cancer Res. 2013;73(17):5315–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Xu C, Li X, Liu P, Li M, Luo F. Patient-derived xenograft mouse models: A high fidelity tool for individualized medicine. Oncol Lett. 2019;17(1):3–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Gao H, Korn JM, Ferretti S, Monahan JE, Wang Y, Singh M, et al. High-throughput screening using patient-derived tumor xenografts to predict clinical trial drug response. Nat Med. 2015;21(11):1318–25. [DOI] [PubMed] [Google Scholar]
  • 122.Cranmer LD, Trevor KT, Bandlamuri S, Hersh EM. Rodent models of brain metastasis in melanoma. Melanoma Res. 2005;15(5):325–56. [DOI] [PubMed] [Google Scholar]
  • 123.Hart IR, Fidler IJ. Cancer invasion and metastasis. Q Rev Biol. 1980;55(2):121–42. [DOI] [PubMed] [Google Scholar]
  • 124.Stephen Paget’s paper reproduced from The Lancet, 1889. Cancer and Metastasis Reviews. 1989;8(2):98–101. [PubMed] [Google Scholar]
  • 125.Lorger M, Felding-Habermann B. Capturing changes in the brain microenvironment during initial steps of breast cancer brain metastasis. Am J Pathol. 2010;176(6):2958–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Contreras-Zarate MJ, Ormond DR, Gillen AE, Hanna C, Day NL, Serkova NJ, et al. Development of Novel Patient-Derived Xenografts from Breast Cancer Brain Metastases. Front Oncol. 2017;7:252. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Bos PD, Nguyen DX, Massague J. Modeling metastasis in the mouse. Curr Opin Pharmacol. 2010;10(5):571–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Cooper JB, Ronecker JS, Tobias ME, Mohan AL, Hillard V, Murali R, et al. Molecular Sequence of Events and Signaling Pathways in Cerebral Metastases. Anticancer Res. 2018;38(4):1859–77. [DOI] [PubMed] [Google Scholar]
  • 129.Singh M, Venugopal C, Tokar T, Brown KR, McFarlane N, Bakhshinyan D, et al. RNAi screen identifies essential regulators of human brain metastasis-initiating cells. Acta Neuropathol. 2017;134(6):923–40. [DOI] [PubMed] [Google Scholar]
  • 130.Mathieu A, Remmelink M, D’Haene N, Penant S, Gaussin JF, Van Ginckel R, et al. Development of a chemoresistant orthotopic human nonsmall cell lung carcinoma model in nude mice: analyses of tumor heterogenity in relation to the immunohistochemical levels of expression of cyclooxygenase-2, ornithine decarboxylase, lung-related resistance protein, prostaglandin E synthetase, and glutathione-S-transferase-alpha (GST)-alpha, GST-mu, and GST-pi. Cancer. 2004;101(8):1908–18. [DOI] [PubMed] [Google Scholar]
  • 131.Kurebayashi J, McLeskey SW, Johnson MD, Lippman ME, Dickson RB, Kern FG. Quantitative demonstration of spontaneous metastasis by MCF-7 human breast cancer cells cotransfected with fibroblast growth factor 4 and LacZ. Cancer Res. 1993;53(9):2178–87. [PubMed] [Google Scholar]
  • 132.Bailey-Downs LC, Thorpe JE, Disch BC, Bastian A, Hauser PJ, Farasyn T, et al. Development and characterization of a preclinical model of breast cancer lung micrometastatic to macrometastatic progression. PLoS One. 2014;9(5):e98624. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Rozenberg GI, Monahan KB, Torrice C, Bear JE, Sharpless NE. Metastasis in an orthotopic murine model of melanoma is independent of RAS/RAF mutation. Melanoma Res. 2010;20(5):361–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Rofstad EK. Orthotopic human melanoma xenograft model systems for studies of tumour angiogenesis, pathophysiology, treatment sensitivity and metastatic pattern. Br J Cancer. 1994;70(5):804–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Lockshin A, Giovanella BC, Stehlin JS Jr. Quantitative evaluation of anticancer agents against human melanoma cells implanted in nude mice. Exp Cell Biol. 1986;54(3):149–54. [DOI] [PubMed] [Google Scholar]
  • 136.Singh SK, Hawkins C, Clarke ID, Squire JA, Bayani J, Hide T, et al. Identification of human brain tumour initiating cells. Nature. 2004;432(7015):396–401. [DOI] [PubMed] [Google Scholar]
  • 137.Ireson CR, Alavijeh MS, Palmer AM, Fowler ER, Jones HJ. The role of mouse tumour models in the discovery and development of anticancer drugs. Br J Cancer. 2019;121(2):101–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Cruz-Munoz W, Kerbel RS. Preclinical approaches to study the biology and treatment of brain metastases. Semin Cancer Biol. 2011;21(2):123–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Arvanitis CD, Ferraro GB, Jain RK. The blood-brain barrier and blood-tumour barrier in brain tumours and metastases. Nat Rev Cancer. 2020;20(1):26–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Zottel A, Videtic Paska A, Jovcevska I. Nanotechnology Meets Oncology: Nanomaterials in Brain Cancer Research, Diagnosis and Therapy. Materials (Basel). 2019;12(10). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Ni J, Miao T, Su M, Khan NU, Ju X, Chen H, et al. PSMA-targeted nanoparticles for specific penetration of blood-brain tumor barrier and combined therapy of brain metastases. J Control Release. 2021;329:934–47. [DOI] [PubMed] [Google Scholar]
  • 142.Wyatt RM, Fraser I, Welty N, Lord B, Wennerholm M, Sutton S, et al. Pharmacologic Characterization of JNJ-42226314, [1-(4-Fluorophenyl)indol-5-yl]-[3-[4-(thiazole-2-carbonyl)piperazin-1-yl]azetidin −1-yl]methanone, a Reversible, Selective, and Potent Monoacylglycerol Lipase Inhibitor. J Pharmacol Exp Ther. 2020;372(3):339–53. [DOI] [PubMed] [Google Scholar]
  • 143.Yin W, Zhao Y, Kang X, Zhao P, Fu X, Mo X, et al. BBB-penetrating codelivery liposomes treat brain metastasis of non-small cell lung cancer with EGFR(T790M) mutation. Theranostics. 2020;10(14):6122–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Mohammad AS, Griffith JI, Adkins CE, Shah N, Sechrest E, Dolan EL, et al. Liposomal Irinotecan Accumulates in Metastatic Lesions, Crosses the Blood-Tumor Barrier (BTB), and Prolongs Survival in an Experimental Model of Brain Metastases of Triple Negative Breast Cancer. Pharm Res. 2018;35(2):31. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Mohsenikia M, Farhangi B, Alizadeh AM, Khodayari H, Khodayari S, Khori V, et al. Therapeutic effects of dendrosomal solanine on a metastatic breast tumor. Life Sci. 2016;148:260–7. [DOI] [PubMed] [Google Scholar]
  • 146.Tahmasebi Mirgani M, Isacchi B, Sadeghizadeh M, Marra F, Bilia AR, Mowla SJ, et al. Dendrosomal curcumin nanoformulation downregulates pluripotency genes via miR-145 activation in U87MG glioblastoma cells. Int J Nanomedicine. 2014;9:403–17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Kinoshita M, McDannold N, Jolesz FA, Hynynen K. Noninvasive localized delivery of Herceptin to the mouse brain by MRI-guided focused ultrasound-induced blood-brain barrier disruption. Proc Natl Acad Sci U S A. 2006;103(31):11719–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Kobus T, Zervantonakis IK, Zhang Y, McDannold NJ. Growth inhibition in a brain metastasis model by antibody delivery using focused ultrasound-mediated blood-brain barrier disruption. J Control Release. 2016;238:281–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Alkins R, Burgess A, Kerbel R, Wels WS, Hynynen K. Early treatment of HER2-amplified brain tumors with targeted NK-92 cells and focused ultrasound improves survival. Neuro Oncol. 2016;18(7):974–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Barth RF, Yang W, Bartus RT, Rotaru JH, Ferketich AK, Moeschberger ML, et al. Neutron capture therapy of intracerebral melanoma: enhanced survival and cure after blood-brain barrier opening to improve delivery of boronophenylalanine. Int J Radiat Oncol Biol Phys. 2002;52(3):858–68. [DOI] [PubMed] [Google Scholar]
  • 151.Gril B, Palmieri D, Bronder JL, Herring JM, Vega-Valle E, Feigenbaum L, et al. Effect of lapatinib on the outgrowth of metastatic breast cancer cells to the brain. J Natl Cancer Inst. 2008;100(15):1092–103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Tomasello G, de Azambuja E, Dinh P, Snoj N, Piccart-Gebhart M. Jumping higher: is it still possible? The ALTTO trial challenge. Expert Rev Anticancer Ther. 2008;8(12):1883–90. [DOI] [PubMed] [Google Scholar]
  • 153.Palmieri D, Lockman PR, Thomas FC, Hua E, Herring J, Hargrave E, et al. Vorinostat inhibits brain metastatic colonization in a model of triple-negative breast cancer and induces DNA double-strand breaks. Clin Cancer Res. 2009;15(19):6148–57. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Yin JJ, Zhang L, Munasinghe J, Linnoila RI, Kelly K. Cediranib/AZD2171 inhibits bone and brain metastasis in a preclinical model of advanced prostate cancer. Cancer Res. 2010;70(21):8662–73. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Klar U, Buchmann B, Schwede W, Skuballa W, Hoffmann J, Lichtner RB. Total synthesis and antitumor activity of ZK-EPO: the first fully synthetic epothilone in clinical development. Angew Chem Int Ed Engl. 2006;45(47):7942–8. [DOI] [PubMed] [Google Scholar]
  • 156.Thomas FC, Taskar K, Rudraraju V, Goda S, Thorsheim HR, Gaasch JA, et al. Uptake of ANG1005, a novel paclitaxel derivative, through the blood-brain barrier into brain and experimental brain metastases of breast cancer. Pharm Res. 2009;26(11):2486–94. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157.Ippen FM, Alvarez-Breckenridge CA, Kuter BM, Fink AL, Bihun IV, Lastrapes M, et al. The Dual PI3K/mTOR Pathway Inhibitor GDC-0084 Achieves Antitumor Activity in PIK3CA-Mutant Breast Cancer Brain Metastases. Clin Cancer Res. 2019;25(11):3374–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Cook D, Brown D, Alexander R, March R, Morgan P, Satterthwaite G, et al. Lessons learned from the fate of AstraZeneca’s drug pipeline: a five-dimensional framework. Nat Rev Drug Discov. 2014;13(6):419–31. [DOI] [PubMed] [Google Scholar]
  • 159.Moreno L, Pearson AD. How can attrition rates be reduced in cancer drug discovery? Expert Opin Drug Discov. 2013;8(4):363–8. [DOI] [PubMed] [Google Scholar]
  • 160.Kerbel RS. Human tumor xenografts as predictive preclinical models for anticancer drug activity in humans: better than commonly perceived-but they can be improved. Cancer Biol Ther. 2003;2(4 Suppl 1):S134–9. [PubMed] [Google Scholar]
  • 161.Mattie M, Christensen A, Chang MS, Yeh W, Said S, Shostak Y, et al. Molecular characterization of patient-derived human pancreatic tumor xenograft models for preclinical and translational development of cancer therapeutics. Neoplasia. 2013;15(10):1138–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Julien S, Merino-Trigo A, Lacroix L, Pocard M, Goere D, Mariani P, et al. Characterization of a large panel of patient-derived tumor xenografts representing the clinical heterogeneity of human colorectal cancer. Clin Cancer Res. 2012;18(19):5314–28. [DOI] [PubMed] [Google Scholar]

RESOURCES