Next Article in Journal
MicroRNA-9-5p-CDX2 Axis: A Useful Prognostic Biomarker for Patients with Stage II/III Colorectal Cancer
Previous Article in Journal
Enhanced Hsa-miR-181d/p-STAT3 and Hsa-miR-181d/p-STAT5A Ratios Mediate the Anticancer Effect of Garcinol in STAT3/5A-Addicted Glioblastoma
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Bioprofiling TS/A Murine Mammary Cancer for a Functional Precision Experimental Model

by
Carla De Giovanni
1,
Giordano Nicoletti
2,
Lorena Landuzzi
2,
Arianna Palladini
1,
Pier-Luigi Lollini
1,*,† and
Patrizia Nanni
1,†
1
Department of Experimental, Diagnostic and Specialty Medicine, Alma Mater Studiorum University of Bologna, Bologna I-40126, Italy
2
Laboratory of Experimental Oncology, IRCCS Istituto Ortopedico Rizzoli, Bologna I-40136, Italy
*
Author to whom correspondence should be addressed.
These authors contributed equally to this paper.
Cancers 2019, 11(12), 1889; https://doi.org/10.3390/cancers11121889
Submission received: 23 October 2019 / Revised: 20 November 2019 / Accepted: 22 November 2019 / Published: 27 November 2019

Abstract

:
The TS/A cell line was established in 1983 from a spontaneous mammary tumor arisen in an inbred BALB/c female mouse. Its features (heterogeneity, low immunogenicity and metastatic ability) rendered the TS/A cell line suitable as a preclinical model for studies on tumor–host interactions and for gene therapy approaches. The integrated biological profile of TS/A resulting from the review of the literature could be a path towards the description of a precision experimental model of mammary cancer.

1. Introduction

Precision medicine in clinics is an evolving concept which goes beyond mere genomic medicine and means matching individual patients with medicine [1]. According to these premises, in an experimental environment a precision cancer model should mean matching the appropriate preclinical model with target biology study [2]. Preclinical models of mammary cancer of increasing complexity have been proposed, including transplantable murine tumors, gene-driven mammary carcinogenic models, human cell lines grown in vitro or in vivo as xenografts and patient-derived xenografts and organoids (see Section 6 for a comparative discussion) [3,4]. Each model remains an approximation [2], with advantages and disadvantages depending on the specific aim of the study. The main advantage of transplantable murine mammary tumors consists of allowing mechanistic studies on tumor–host interactions, like those focusing on the role of microenvironment, the metastatic process and the immune response. A deep knowledge of a preclinical model, where literature studies are collected and retrospectively examined as a whole, like an individual patient’s medical record, can help in a better design of experimental approaches. The aim of this review is the biological profiling of a popular model of murine mammary cancer (TS/A) for a better understanding and modeling of a complex pathology like human breast cancer [5].

2. The Dawn of Murine Models for Tumor–Host Interactions

At the beginning of the 1980s, metastases and tumor–host interaction studies mostly took advantage of a few tumor cell lines, established and subcultured for many years, such as 3LL Lewis lung carcinoma and B16 murine spontaneous melanoma [6]. Through the intravenous injection of B16 cells, metastatic deposits to lungs and other organs could be easily obtained, allowing for important advancements in understanding post-intravasation late phases of the metastatic process. However, the B16 parental cell line was almost incapable of disseminating from a locally-growing tumor, and therefore it did not adequately model the invasion and intravasation phases. Moreover, the non-epithelial origin of B16 melanoma impeded inferences about the behavior of epithelial tumors. At the same time, some rodent cell lines were already being used as models for mammary cancer, but most of them were either carcinogen- or virus-induced [7,8]. These models did not undergo a long natural history in the host, in which they arose, and generally had a high immunogenicity due to the expression of strong tumor-associated antigens. Likely due to these features, they generally gave too optimistic results when used to study antitumor immune responses or immunotherapeutic approaches [9].
In this landscape, in 1983 we described a new cell line, TS/A, derived from a mammary tumor spontaneously arisen in a 20 month-old BALB/c inbred mouse strain [10]. The TS/A cell line exhibited some features typical of human breast cancer, which prompted its use as a preclinical model, such as the low immunogenicity, the ability of local tumors to give rise to distant metastases and the heterogeneity, well evident both of morphology and metastatic ability. The TS/A cell line (also referred to as TSA or TS/A-pc, see [11,12] and below) and its clones were distributed to many laboratories worldwide and were employed for different applications, such as studies on malignant phenotype, pharmacologic therapeutic approaches, antitumor immune response and as a gene therapy model.
A list of research studies exploiting the TS/A cell line or its cell variants is reported in the Supplementary Table S1. It includes (up to 2018) 276 research papers where TS/A was used as model system and 19 papers where it was a control model. Reviews reporting results obtained with TS/A and citations of the TS/A paper are also listed in the Supplementary Table S1.
This review aims at profiling the main biological features of the TS/A model system resulting from literature research papers (Table 1). The two research areas where TS/A-based models yielded important results will be discussed in depth: tumor–host interactions and experimental gene therapy.

3. Bioprofiling TS/A Cell Line

The mammary tumor originating the TS/A cell line was isolated in a 20 months-old BALB/c female retired breeder and was described as a moderately differentiated adenocarcinoma [10]. Its first in vivo passage into a healthy BALB/c female was adapted to in vitro culture and named TS/A (Figure 1). Several clones and cell variants were derived from TS/A and distributed worldwide. In particular, a TS/A subline was chosen by Guido Forni (University of Turin) for a large collaborative endeavor as the recipient cell for the systematic transduction of a large series of genes coding for immune modulators; such subline was referred to as TS/A-pc (from “parental cells”). TS/A and TS/A-pc share most features reviewed here, and some kind of drift occurred during the extensive amplification of TS/A-pc. Throughout this review, we will refer to the TS/A model system on the whole, and therefore incorrect terminology (such as TSA, TS/a, and so on) has been systematically corrected to “TS/A”. However, the Supplementary Table reports exactly the TS/A cell variant used in each referenced paper.
The tumor from which the TS/A cell line was derived likely had a long natural history in its host of origin. When tested in a growth-excision test, TS/A cells did not confer protection against a second challenge, thus showing a low immunogenicity [10], thereafter confirmed in other studies (see, for example, [23]). Such a low immunogenicity was the basis for a huge number of immunopotentiation studies, most of which exploiting gene therapy approaches (see next section).
TS/A cells express the gp70env product of an endogenous retrovirus whose AH1 immunodominant class I epitope could be recognized by cytotoxic T lymphocytes through presentation by H-2Ld [25]. Gp70env antigen is shared by other murine cell lines, such as the colorectal cancer cell line CT26. The down-regulation of the Ld observed in TS/A cells [52] is likely due to the immunoediting process leading to evasion from the host immune response.
TS/A exerts a suppressive effect on the host immune response through several mechanisms, such as a selective loss of STAT5a/b expression in T and B lymphocytes [26], the production of transforming growth factor β1 (TGF-β1) [18], the induction of regulatory T cells [27], natural killer resistance [36] and the production of colony stimulating factors (CSFs) that deeply subvert hematopoiesis [14,15], giving rise to splenomegaly, leucocytosis and to tumor-infiltrating myeloid-derived suppressor cells (MDSC) [29,33,53].
When injected subcutaneously into syngeneic BALB/c mice, TS/A cells gave rise to local tumors rapidly disseminating to the lungs. Metastases could also be obtained after injection of TS/A cells by the intravenous route, thus allowing a comparison between the dynamics of the early and late phases of the metastatic process [10,41]. Metastases to lungs and liver have also been obtained by orthotopic cell injection [54]. Like other mammary carcinoma cell lines, the growth of micrometastases at distant organs was found to involve the formation of filopodium-like protrusions mediated by FAK/ERK and Rif/mDia2 signaling [54].
Heterogeneity of TS/A cells was observed in adherent cultures, with areas of epithelial-like and fibroblast-like morphology (Figure 2), and in anchorage-independent cultures [10,40]. Subcloning from agar cultures allowed the isolation of two types of cell clones, both tumorigenic and metastatic, but with markedly different metastatic power [40]. Unexpectedly high-metastatic clones had a prevalent epithelial morphology, compared to the fibroblast-like pattern of low-metastatic clones. Gene expression profiling of several murine mammary cancer cell lines showed clustering of TS/A-E1 (a high-metastatic clone) with high-claudin expressors [13]. Our data on gene expression profiling of TS/A clones showed that claudin-3 was the top overexpressed gene in high-metastatic clones (about 90-fold expression over low-metastatic clones), while low-metastatic clones overexpressed nme4 and necdin, two putative metastasis suppressor genes (our unpublished results). These data suggest that metastatic ability is not always a consequence of epithelial–mesenchymal transition but can also be acquired in an epithelial-like differentiation context.
The TS/A cell line has a triploid karyotype [20] and carries a mutated p53 at codon 270 [21]. About a third of the cells express the cancer stem cell marker Sca-1 [43]. In our laboratory, the expression of Sca-1 (also known as Ly6A) was almost negative, but inducible by IFN-γ [12]. TS/A cells express estrogen receptor [10] and endogenous murine p185-erbB2 product [39]. Its use as a negative HER2/neu mammary cancer cell line in studies on HER2/neu transgenic models relies upon the negativity to the reagent specifically recognizing rat HER2/neu.

4. Tumor–Host Interaction Studies

TS/A-induced tumors have a rich and heterogeneous infiltrate comprising granulocyte and monocyte/macrophage subpopulations, whose relative proportions change during tumor progression [55], in agreement with the known plasticity of myeloid cells. Several subpopulations contribute to maintainance of a tumor-promoting microenvironment in TS/A, as well as in many other murine and human tumors [56], with a variety of mechanisms. Alternatively-activated M2 macrophages are strong producer of the immunosuppressive cytokine IL10 and of several chemokines recruiting Treg, Th2, eosinophils and basophils [56]. MDSC are heterogeneous immature CD11b+/Gr-1+ populations [57], with immunosuppressive function. Both M2-polarized macrophages and MDSC have been investigated in the TS/A model system, along with strategies to circumvent tumor promotion, pushing infiltrate cells towards more differentiated, activated cells.
In the TS/A model, the induction of M2 tumor-associated macrophages was mediated by the expression of the CD20 homolog MS4A8A gene [58]. In TS/A tumors, M2-polarized macrophages were more abundant and more proangiogenic in hypoxic tumor areas [55]. The M2 immune suppressing phenotype was switched to an anti-tumor M1 phenotype through the in vivo adenoviral gene transfer of the chemokine CCL16 [59]. Alternatively-activated M2 macrophages expressed highly restricted, individual-specific, combinatorial T cell receptor-αβ immunoreceptors, suggestive of an adaptive response of macrophages to the tumor [60]. In TS/A, as well as in a variety of other murine and human tumors, alternatively-activated M2 tumor-associated macrophages expressed a multifunctional scavenger receptor named stabilin-1 involved in endocytic and phagocytic clearance of “unwanted-self” components, including soluble component of extracellular matrix SPARC (a tumor-inhibiting agent). Stabilin-1 was found to play a tumor promoting role in the TS/A model likely through enhanced clearance of SPARC [61].
A major component of TS/A infiltrate consisted of MDSC [53], which correlated with the production of CSFs by TS/A cells [14,29]. Immature myeloid progenitors can be released in the bloodstream, giving rise to peripheral leukocytosis and splenomegaly [14,15]. MDSC suppressed antigen-activated T lymphocytes through apoptosis induction [28,29], and suppressed NK cytotoxicity [62], with mechanisms involving nitric oxide [30]. Impaired anti-tumor immune response in aging can take advantage of an increased MDSC infiltrate [32]. MDSC expressed Fas–FasL and caspases, suggesting that Fas–FasL apoptosis regulated MDSC survival [33,34] and proposing new potential therapeutic options. MDSCs are key drivers of resistance to antiangiogenic therapy, but all-trans retinoic acid was able to induce differentiation of MDSC into mature cells, thus increasing the efficacy of the antiangiogenic therapy [63].
In the TS/A microenvironment, other non-tumoral cell types can play a tumor-promoting role, such as tumor-associated fibroblasts and adipocytes. Through a tumor-stromal cell co-injection model, novel candidate tumor-associated genes were identified in tumor-associated fibroblasts. The most studied gene was tubulin tyrosine ligase: its downregulation in tumor-associated fibroblasts promoted TS/A tumor growth [64]. Co-culture of TS/A cells with adipocytes caused an increased lipid content in TS/A cells and an increased lung colonization ability [65]. The release of free fatty acids from lipid droplets is mediated by an adipose triglyceride lipase-dependent lipolytic pathway, that was proposed as a potential therapeutic target. The metabolic cross-talk between tumor cells and tumor-associated adipocytes could favor epithelial-mesenchymal transition and increase tumor invasiveness.
TS/A cells, like other tumor cell lines, secrete membrane vesicles of endosomal origin called “exosomes”, with contradictory roles in tumor biology. Exosomes could have some immunostimulatory effect, since they carry tumor antigens which can be transferred to dendritic cells and cross-prime cytotoxic T lymphocytes [66]. However, exosomes mainly exerted a potent immunosuppressive anti-tumor immune response through suppression of NK cell function [67] and inhibition of differentiation of bone marrow dendritic cells [68]. Tumor-derived exosomes released from irradiated TS/A cells showed an altered molecular composition and were able to transfer dsDNA to dendritic cells and stimulate upregulation of costimulatory molecules and STING-dependent activation of IFN-I [69].

5. Gene Therapy Studies

TS/A cells were easily transduced both with naked DNA and viral systems, generating good and stable transgene expression of secreted factors or membrane molecules. Most gene therapy approaches were performed to directly increase TS/A immunogenicity with the purpose to use engineered cells as anticancer vaccines (Table 2).
Genes for a variety of cytokines, costimulatory molecules and major histocompatibility complex (MHC) antigens were inserted and stably expressed in TS/A cells. Cytokine transduction in TS/A cells was often performed isolating clones with different levels of cytokine production, and this allowed to study the dose-related effects, such as the minimal cytokine release level required to significantly impact on tumor growth and immunogenicity and the potential side effects of highly-releasing cells. As an example, IFN-γ transduction led to isolate clones with cytokine production ranging from a few IU/ml up to a very high expressor clone (releasing 6000 IU/mL), likely the highest transduced expression ever obtained. Such a panel of IFN-γ releasing clones showed a dose-related growth inhibition and immunogenicity, but also showed potentially important side effects, such as increased lung colonizing ability and other systemic effects [12,136].
The wide portfolio of TS/A cells transduced with different cytokine genes allowed to understand the role played by each cytokine in the modulation of tumor infiltrate composition and its impact on tumor growth [137]. A major role for granulocytes in cytokine-induced tumor debulking was unexpectedly found, along with a continuous cross-talk between leukocytes and lymphocytes. The transduced cytokine drove the composition of the reactive cells elicited, the efficacy of the anti-tumor reaction and the immune memory against the non-transduced tumor. The increased memory reaction is the basis for the use of gene-engineered cells as anticancer vaccines. On the whole, data obtained with engineered TS/A vaccines (Table 2) showed that the most effective cytokines were IFN-γ and IL-12.
TS/A transduction with GM-CSF was performed only once [74], with almost no effect on tumor growth or immunogenicity. On the contrary, GM-CSF engineering of another murine model (B16 melanoma) gave good results [138] and prompted clinical studies. B16 melanoma did not produce spontaneously GM-CSF whereas TS/A abundantly secreted CSFs [16]. The spontaneous CSF production in TS/A did not hamper tumor growth but likely contributed to the tumor-promoting environment, showing that similar cytokines could play opposite roles in tumors of different origin.
Transduction of genes coding for activating pro-drug enzymes (suicide genes) was performed with the main aim to obtain more immunogenic cancer cell vaccines. It was reported that replicating cells were more immunogenic than dead cells [74], so prodrug activation by suicide gene products could switch off partially replicating cell vaccines after the start of the immune response. However, prodrug-induced cancer cell death itself was found to increase the specific immune response [125]. Suicide genes were also included in oncolytic viruses, to enhance their safety profile [128].
Gene therapy approaches to obtain increased TS/A cancer cell immunogenicity gave interesting but, at the same time, unsatisfactory results. Most approaches actually showed increased immunogenicity, but when challenged in therapeutic set up, a minority of mice could be cured, and only when therapy started at the very early phases of metastatic growth [106,112]. Similar conclusions could be drawn for the variety of gene therapy trials conducted in the last three decades with the purpose of increasing tumor immunogenicity through cytokine or costimulatory gene transduction. Therefore, results obtained with TS/A as well as with other experimental gene therapy models predicted the low efficacy found in trials. Combined gene therapy approaches showed better therapeutic activity and prompted new combination immune-gene therapy approaches [99,111].
Gene transduction was applied to the TS/A model to study cancer biology and cancer gene therapy (Table 3). Transduction of the wild-type p53 gene (p53wt), aiming to restore a correct p53 signaling, was performed in vitro and in vivo with a Canarypox vector carrying p53wt, leading to downstream p21 expression with a proapoptotic effect that caused tumor growth inhibition [21]. Tumor rejection was associated with the generation of a specific antitumor immune response in a sarcoma model but not in TS/A, thus confirming the low immunogenicity of the TS/A model system.
TS/A cells were transduced with luciferase gene and green fluorescent protein (GFP) variants and used in studies on imaging techniques (Table 3). TS/A cells were used as recipient for genes coding exogenous antigens as a surrogate to study features of the corresponding immune response (Table 3).
Silencing approaches were performed with retro- and lenti-viral vectors and recently with CRISPR-Cas9 technology. Through silencing, TGF-β1 released by TS/A cells was found to play a suppressive role on graft-versus-tumor reaction [18].
In search of new genes potentially involved in metastasis of mammary cancer, along with data from human histopathological samples, some studies used TS/A cells for a mechanistic demonstration through silencing approaches. These studies were sometimes performed in parallel with another popular model of murine mammary cancer (4T1), which is more metastatic than TS/A cells (see Section 6). The overexpression of Fragile X mental retardation protein (FMRP) was concordantly related to lung metastases in both models [46]. On the contrary, some disagreement between TS/A and 4T1 was reported concerning the role of the small GTPase Rab27a [146]. Rab27a was involved in exosome secretion. Its silencing inhibited tumor growth and lung metastases in the 4T1 model, but not in TS/A. It should be noted that the authors described TS/A as a non-metastatic tumor model. Since TS/A is actually able to metastasize to lungs, two explanations are possible for such discrepancy: a) Rab27a is not an on/off determinant of metastatic power, but rather a quantitative modulator; b) 4T1 is a clone while TS/A is a polyclonal and heterogeneous cell line. TS/A extensive subculture can have led to drift phenomena with oligoclonal dominance of less metastatic cells, which are well represented in the cell line of origin.
Genetic inactivation through CRISPR/CAS9 technology of the DNA mismatched repair gene MutL homologue 1 (MLH1) in TS/A cells, as well as in other non-mammary murine cancer models, led to increased immunogenicity due to accumulation of neoantigens [147]. MLH1-inactivated cells acquired sensitivity to antibodies against checkpoint inhibitors, which now represent the forefront of cancer immunotherapy.
The expression of murine ErbB2 in TS/A cells was exploited to provide experimental evidence of the oncosuppressor role of FoxP3 in mammary cancers, that downmodulated the expression of the ErbB2 oncogene [44]. TS/A cells was also used as a model to study optimization of parameters of gene electrotransfer [50].

6. Comparison with Other Mammary Cancer Models

Modeling mammary cancer in mouse to study tumor–host interactions took advantage of several model systems [3,4,166]. Reordering models according to their intrinsic complexity, we can mention transplantable murine tumors, gene-driven mammary carcinogenic models, human cell lines grown in vitro or in vivo as xenografts and patient-derived xenografts and organoids.
Concerning transplantable murine mammary cancer, the most popular cell line is 4T1, derived from a spontaneous mammary cancer arisen in a BALB/cfC3H female [167,168,169]. 4T1 share several features with TS/A and with human mammary cancers, such as low immunogenicity and tumor–host interactions. In fact, several studies were performed using in parallel 4T1 and TS/A (see for example Supplementary Table S1, column N), which were considered as biological replicates and generally gave concordant results. We can focus here on the main differences between the two models. 4T1 is a thioguanine-resistant clone derived from a heterogeneous mammary cancer cell line [167,168]. TS/A is a cell line with heterogeneity spanning from morphology to metastatic ability and to CSF production (and therefore tumor–host interactions), as proven by the in vitro isolation of clones with markedly different features [14,40]. Populations with different abilities to metastasize were also isolated from TS/A through in vivo selection procedures [41]. Heterogeneity is a hallmark of mammary cancer, which comprises morphology, differentiation and metastatic ability, but cloned populations at least partially lose such heterogeneity. 4T1 is a highly aggressive clone, with the ability to give rise to a high number of lung metastases following subcutaneous, intravenous or orthotopic cell injections (see for example [146]). Moreover, 4T1 can metastasize to other organs (such as liver and bone) [170]. TS/A is a cell line provided with metastatic ability but giving rise to moderate number of lung metastases following local growth, subcutaneous and intravenous injections or orthotopic administration [10,54]. The lower metastatic ability of the TS/A model can allow to study a wider range of metastasis modulators.
In the last three decades the research on mammary tumor development and malignancy took advantage of transgenic models. One of the most studied models of gene-driven mammary carcinogenesis was that based on the rat HER2/neu oncogene under the transcriptional control of the Mouse Mammary Tumor Virus (MMTV) promoter [171,172]. Transgenic models recapitulated all the transitions from the normal mammary gland to mammary cancer, both from morphological and molecular points of view, and led to essential advancement in comprehension of the carcinogenic process and development of new therapeutic approaches. The reproducible carcinogenic process observed in transgenic models was exploited to study the prevention of tumor progression, including approaches based on immune strategies [173]. Transgene expression is somewhat artificial, concerning both the xenogeneic origin of the oncogene (rat HER2/neu) and the expression driven by a viral promoter. From an immunological point of view, the fast carcinogenesis and the altered immunoreactivity of mice being tolerant to transgene can be significant differences from the human pathogenetic development of mammary cancer. However, the main problems of transgenic models are time-consuming procedures and costs. Cell lines from spontaneous mammary cancer such as TS/A therefore are still widely employed in studies on biological features and new therapeutic approaches.
Human models for mammary cancer comprise cell lines and xenografts [4,166]. Human breast cancer is a heterogeneous disease that, thanks to biomarkers, can be subdivided in different subtypes with prognostic significance [174,175] and subjected to appropriate treatments. The main advantage of human models is that they can reproduce the heterogeneity among tumors, giving researchers the possibility to choose the correct subtype depending on the aim of the research, while the main constraint of human cell lines and xenografts is the lack of immune tumor–host interactions. To identify which subtypes can be modeled by the different murine mammary cancers, a comparison among gene expression profiles of a panel of murine mammary cancer cell lines including a TS/A variant (clone E1) and profiles of the different human subtypes was performed [13]. E1 showed a non-basal profile, with prevalent features of luminal A and HER2 subtypes (about 50% and 20%–30% probability, respectively). Therefore, a single murine model can mimic a peculiar human subtype, but obviously is limited to the fixed genetic setting of the cell line and does not reflect diverse spectrum of personalized genetic and/or epigenetic alterations of human breast cancers.
To better depict individual mammary cancers, patient-derived xenografts (PDX) [176] and patient-derived organoids (PDO) were proposed [177,178,179]. Such approaches are more compatible with the need of precision oncology and without concern of species difference. PDX do not allow to study immune interactions (since they are grown in immunodeficient mice), and also present other disadvantages such as the low frequency of tumor take, with a bias toward more aggressive subtypes [180], the cost and the time-consuming procedure. PDO are 3D cultures obtained by dissociated tumor tissue which can be co-cultured with human lymphocytes, thus allowing to investigate tumor microenvironment, anticancer immunotherapy, and other aspects including development of novel therapeutics [181,182].
In conclusion, preclinical models of murine mammary cancer cell lines are still widely used thanks to their possibility to focus on tumor–host interactions comprising the role of stroma, the metastatic process and immune responses. The recent burst of immune-based anti-cancer therapies (see for example checkpoint inhibitors [133,147] and CAR-T [183]) likely will take advantage of murine models comprising mammary cancer cell lines. Other advantages are the low cost and time to obtain results. The possibility to study in parallel several cancer cell lines mimicking different breast cancer subtypes could remain a first-line means to study innovative molecular and therapeutic approaches, which will be then tested in individually precise, more complex human models.

7. Conclusions

The analysis of the main studies exploiting TS/A as a pre-clinical model of mammary cancer allows to draft a profile spanning from molecular alterations to malignant phenotype and immune interactions. This profile should be considered when designing experiments based on TS/A model. Knowledge of this profile can allow inference about the complexity of human breast cancer.

Supplementary Materials

The following are available online at https://www.mdpi.com/2072-6694/11/12/1889/s1, Table S1: Comprehensive list of papers using TS/A cell variants.

Author Contributions

Conceptualization: C.D.G., P.-L.L. and P.N.; Data collection and writing-original draft preparation: C.D.G. and A.P.; Discussion, writing-review and editing: C.D.G., G.N., L.L., A.P., P.-L.L. and P.N.

Funding

This research was funded by grants from the Italian Association for Cancer Research (AIRC) (IG15324 to P-L. Lollini), the Department of Experimental, Diagnostic and Specialty Medicine of the University of Bologna (DIMES) (“Pallotti” Fund) and the University of Bologna.

Conflicts of Interest

The University of Bologna granted to EMD Millipore license for TS/A distribution worldwide. Royalties are destined for oncological research.

References

  1. Letai, A. Functional precision cancer medicine-moving beyond pure genomics. Nat. Med. 2017, 23, 1028–1035. [Google Scholar] [CrossRef] [PubMed]
  2. Gould, S.E.; Junttila, M.R.; De Sauvage, F.J. Translational value of mouse models in oncology drug development. Nat. Med. 2015, 21, 431–439. [Google Scholar] [CrossRef] [PubMed]
  3. Heppner, G.H.; Miller, F.R.; Shekhar, P.V.M. Nontransgenic models of breast cancer. Breast Cancer Res. 2000, 2, 331–334. [Google Scholar] [CrossRef] [PubMed]
  4. Gengenbacher, N.; Singhal, M.; Augustin, H.G. Preclinical mouse solid tumour models: Status quo, challenges and perspectives. Nat. Rev. Cancer 2017, 17, 751–765. [Google Scholar] [CrossRef]
  5. Haynes, B.; Sarma, A.; Nangia-Makker, P.; Shekhar, M.P. Breast cancer complexity: Implications of intratumoral heterogeneity in clinical management. Cancer Metastasis Rev. 2017, 36, 547–555. [Google Scholar] [CrossRef] [PubMed]
  6. Poste, G.; Doll, J.; Fidler, I.J. Interactions among clonal subpopulations affect stability of the metastatic phenotype in polyclonal populations of B16 melanoma cells. Proc. Natl. Acad. Sci. USA 1981, 78, 6226–6230. [Google Scholar] [CrossRef]
  7. Ramshaw, I.A.; Badenoch-Jones, P. Studies on rat mammary adenocarcinomas: A model for metastasis. Cancer Metastasis Rev. 1985, 4, 195–208. [Google Scholar] [CrossRef]
  8. Price, J.E.; Carr, D.; Tarin, D. Spontaneous and induced metastasis of naturally occurring tumors in mice: Analysis of cell shedding into the blood. J. Natl. Cancer Inst. 1984, 73, 1319–1326. [Google Scholar]
  9. Hewitt, H.B. Second point: Animal tumor models and their relevance to human tumor immunology. J. Biol. Response Mod. 1983, 2, 210–216. [Google Scholar]
  10. Nanni, P.; De Giovanni, C.; Lollini, P.L.; Nicoletti, G.; Prodi, G. TS/A: A new metastasizing cell line from a BALB/c spontaneous mammary adenocarcinoma. Clin. Exp. Metastasis 1983, 1, 373–380. [Google Scholar] [CrossRef]
  11. Cavallo, F.; Di Pierre, F.; Giovarelli, M.; Gulino, A.; Vacca, A.; Stoppacciaro, A.; Forni, M.; Modesti, A.; Forni, G. Protective and Curative Potential of Vaccination with Interleukin-2-Gene-transfected Cells from a Spontaneous Mouse Mammary Adenocarcinoma. Cancer Res. 1993, 53, 5067–5070. [Google Scholar] [PubMed]
  12. Lollini, P.-L.; Bosco, M.C.; Cavallo, F.; De Giovanni, C.; Giovarelli, M.; Landuzzi, L.; Musiani, P.; Modesti, A.; Nicoletti, G.; Palmieri, G.; et al. Inhibition of tumor growth and enhancement of metastasis after transfection of the γ-interferon gene. Int. J. Cancer 1993, 55, 320–329. [Google Scholar] [CrossRef] [PubMed]
  13. Yang, Y.; Yang, H.H.; Hu, Y.; Watson, P.H.; Liu, H.; Geiger, T.R.; Anver, M.R.; Haines, D.C.; Martin, P.; Green, J.E.; et al. Immunocompetent mouse allograft models for development of therapies to target breast cancer metastasis. Oncotarget 2017, 8, 30621–30643. [Google Scholar] [CrossRef] [PubMed]
  14. Nicoletti, G.; Brambilla, P.; De Giovanni, C.; Lollini, P.L.; Del Re, B.; Marocchi, A.; Mocarelli, P.; Prodi, G.; Nanni, P. Colony-stimulating activity from the new metastatic TS/A cell line and its high- and low-metastatic clonal derivatives. Br. J. Cancer 1985, 52, 215–222. [Google Scholar] [CrossRef]
  15. Nicoletti, G.; Lollini, P.L.; Bagnara, G.P.; De Giovanni, C.; Del Re, B.; Bons, L.; Prodi, G.; Nanni, P. Are colony-stimulating factor-producing cells facilitated in the metastatic process? Anticancer Res. 1987, 7, 695–700. [Google Scholar] [PubMed]
  16. Nicoletti, G.; De Giovanni, C.; Lollini, P.L.; Bagnara, G.P.; Scotlandi, K.; Landuzzi, L.; Del Re, B.; Zauli, G.; Prodi, G.; Nanni, P. In vivo and in vitro production of haemopoietic colony-stimulating activity by murine cell lines of different origin: A frequent finding. Eur. J. Cancer Clin. Oncol. 1989, 25, 1281–1286. [Google Scholar] [CrossRef]
  17. Bronte, V.; Chappell, D.B.; Apolloni, E.; Cabrelle, A.; Wang, M.; Hwu, P.; Restifo, N.P. Unopposed production of granulocyte-macrophage colony-stimulating factor by tumors inhibits CD8+ T cell responses by dysregulating antigen-presenting cell maturation. J. Immunol. 1999, 162, 5728–5737. [Google Scholar]
  18. Kummar, S.; Ishii, A.; Yang, H.K.; Venzon, D.J.; Kim, S.J.; Gress, R.E. Modulation of graft-versus-tumor effects in a murine allogeneic bone marrow transplantation model by tumor-derived transforming growth factor-β1. Biol. Blood Marrow Transplant. 2001, 7, 25–31. [Google Scholar] [CrossRef]
  19. Cofano, F.; Fassio, A.; Cavallo, G.; Landolfo, S. Binding of murine 125I-labelled natural interferon-gamma to murine cell receptors. J. Gen. Virol. 1986, 67, 1205–1206. [Google Scholar] [CrossRef]
  20. Jentsch, I.; Geigl, J.; Klein, C.A.; Speicher, M.R. Seven-fluorochrome mouse M-FISH for high-resolution analysis of interchromosomal rearrangements. Cytogenet. Genome Res. 2003, 103, 84–88. [Google Scholar] [CrossRef]
  21. Odin, L.; Favrot, M.; Poujol, D.; Michot, J.P.; Moingeon, P.; Tartaglia, J.; Puisieux, I. Canarypox virus expressing wild type p53 for gene therapy in murine tumors mutated in p53. Cancer Gene Ther. 2001, 8, 87–98. [Google Scholar] [CrossRef] [PubMed]
  22. Yamano, T.; Kaneda, Y.; Hiramatsu, S.H.; Huang, S.; Tran, A.N.; Giuliano, A.E.; Hoon, D.S.B. Immunity against breast cancer by TERT DNA vaccine primed with chemokine CCL21. Cancer Gene Ther. 2007, 14, 451–459. [Google Scholar] [CrossRef] [PubMed]
  23. Cavallo, F.; Giovarelli, M.; Gulino, A.; Vacca, A.; Stoppacciaro, A.; Modesti, A.; Forni, G. Role of neutrophils and CD4+T lymphocytes in the primary and memory response to nonimmunogenic murine mammary adenocarcinoma made immunogenic by IL-2 gene. J. Immunol. 1992, 149, 3627–3635. [Google Scholar]
  24. Rakhmilevich, A.L.; Janssen, K.; Hao, Z.; Sondel, P.M.; Yang, N.S. Interleukin-12 gene therapy of a weakly immunogenic mouse mammary carcinoma results in reduction of spontaneous lung metastases via a T-cell- independent mechanism. Cancer Gene Ther. 2000, 7, 826–838. [Google Scholar] [CrossRef] [PubMed]
  25. Rosato, A.; Dalla Santa, S.; Zoso, A.; Giacomelli, S.; Milan, G.; Macino, B.; Tosello, V.; Dellabona, P.; Lollini, P.L.; De Giovanni, C.; et al. The cytotoxic T-lymphocyte response against a poorly immunogenic mammary adenocarcinoma is focused on a single immunodominant class I epitope derived from the gp70 Env product of an endogenous retrovirus. Cancer Res. 2003, 63, 2158–2163. [Google Scholar] [PubMed]
  26. Pericle, F.; Sconocchia, G.; Segal, D.M.; Bronte, V.; Kirken, R.A.; Dasilva, L. Immunocompromised tumor-bearing mice show a selective loss of STAT5a/b expression in T and B lymphocytes. J. Immunol. 1997, 159, 2580–2585. [Google Scholar] [PubMed]
  27. Piconese, S.; Valzasina, B.; Colombo, M.P. OX40 triggering blocks suppression by regulatory T cells and facilitates tumor rejection. J. Exp. Med. 2008, 205, 825–839. [Google Scholar] [CrossRef]
  28. Apolloni, E.; Bronte, V.; Mazzoni, A.; Serafini, P.; Cabrelle, A.; Segal, D.M.; Young, H.A.; Zanovello, P. Immortalized Myeloid Suppressor Cells Trigger Apoptosis in Antigen-Activated T Lymphocytes. J. Immunol. 2000, 165, 6723–6730. [Google Scholar] [CrossRef]
  29. Bronte, V.; Apolloni, E.; Cabrelle, A.; Ronca, R.; Serafini, P.; Zamboni, P.; Restifo, N.P.; Zanovello, P. Identification of a CD11b+/Gr-1+/CD31+ myeloid progenitor capable of activating or suppressing CD8+ T cells. Blood 2000, 96, 3838–3846. [Google Scholar] [CrossRef]
  30. Mazzoni, A.; Bronte, V.; Visintin, A.; Spitzer, J.H.; Apolloni, E.; Serafini, P.; Zanovello, P.; Segal, D.M. Myeloid Suppressor Lines Inhibit T Cell Responses by an NO-Dependent Mechanism. J. Immunol. 2002, 168, 689–695. [Google Scholar] [CrossRef]
  31. Serafini, P.; Meckel, K.; Kelso, M.; Noonan, K.; Califano, J.; Koch, W.; Dolcetti, L.; Bronte, V.; Borrello, I. Phosphodiesterase-5 inhibition augments endogenous antitumor immunity by reducing myeloid-derived suppressor cell function. J. Exp. Med. 2006, 203, 2691–2702. [Google Scholar] [CrossRef] [PubMed]
  32. Grizzle, W.E.; Xu, X.; Zhang, S.; Stockard, C.R.; Liu, C.; Yu, S.; Wang, J.; Mountz, J.D.; Zhang, H.G. Age-related increase of tumor susceptibility is associated with myeloid-derived suppressor cell mediated suppression of T cell cytotoxicity in recombinant inbred BXD12 mice. Mech. Ageing Dev. 2007, 128, 672–680. [Google Scholar] [CrossRef] [PubMed]
  33. Sinha, P.; Chornoguz, O.; Clements, V.K.; Artemenko, K.A.; Zubarev, R.A.; Ostrand-Rosenberg, S. Myeloid-derived suppressor cells express the death receptor Fas and apoptose in response to T cell-expressed FasL. Blood 2011, 117, 5381–5390. [Google Scholar] [CrossRef] [PubMed]
  34. Ostrand-Rosenberg, S.; Sinha, P.; Chornoguz, O.; Ecker, C. Regulating the suppressors: Apoptosis and inflammation govern the survival of tumor-induced myeloid-derived suppressor cells (MDSC). Cancer Immunol. Immunother. 2012, 61, 1319–1325. [Google Scholar] [CrossRef]
  35. Sinha, P.; Parker, K.H.; Horn, L.; Ostrand-Rosenberg, S. Tumor-induced myeloid-derived suppressor cell function is independent of IFN-γ and IL-4Rα. Eur. J. Immunol. 2012, 42, 2052–2059. [Google Scholar] [CrossRef] [Green Version]
  36. Morandi, B.; Mortara, L.; Chiossone, L.; Accolla, R.S.; Mingari, M.C.; Moretta, L.; Moretta, A.; Ferlazzo, G. Dendritic cell editing by activated natural killer cells results in a more protective cancer-specific immune response. PLoS ONE 2012, 7, e39170. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Mirshahidi, S.; Kramer, V.G.; Whitney, J.B.; Essono, S.; Lee, S.; Dranoff, G.; Anderson, K.S.; Ruprecht, R.M. Overlapping synthetic peptides encoding TPD52 as breast cancer vaccine in mice: Prolonged survival. Vaccine 2009, 27, 1825–1833. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  38. Clément, M.; Rocher, J.; Loirand, G.; Le Pendu, J. Expression of sialyl-Tn epitopes on β1 integrin alters epithelial cell phenotype, proliferation and haptotaxis. J. Cell Sci. 2004, 117, 5059–5069. [Google Scholar] [CrossRef] [Green Version]
  39. Hsu, H.C.; Li, L.; Zhang, H.G.; Mountz, J.D. Genetic regulation of thymic involution. Mech. Ageing Dev. 2005, 126, 87–97. [Google Scholar] [CrossRef]
  40. Lollini, P.L.; De Giovanni, C.; Eusebi, V.; Nicoletti, G.; Prodi, G.; Nanni, P. High-metastatic clones selected in vitro from a recent spontaneous BALB/c mammary adenocarcinoma cell line. Clin. Exp. Metastasis 1984, 2, 251–259. [Google Scholar] [CrossRef]
  41. Nanni, P.; De Giovanni, C.; Lollini, P.L.; Nicoletti, G.; Prodi, G. Clones with different metastatic capacity and variant selection during metastasis: A problematic relationship. J. Natl. Cancer Inst. 1986, 76, 87–93. [Google Scholar] [PubMed]
  42. Lollini, P.L.; Landuzzi, L.; Nlcoletti, G.; De Giovanni, C.; Glovarelli, M.; Lalli, E.; Facchini, A.; Nanni, P. LY-6A/E gene is widely expressed among transformed nonhematopoietic cells. Autocrine modulation by interferon. Anticancer Res. 1992, 12, 2245–2252. [Google Scholar] [PubMed]
  43. Kim, R.J.; Kim, S.R.; Roh, K.J.; Park, S.B.; Park, J.R.; Kang, K.S.; Kong, G.; Tang, B.; Yang, Y.A.; Kohn, E.A.; et al. Ras activation contributes to the maintenance and expansion of Sca-1pos cells in a mouse model of breast cancer. Cancer Lett. 2010, 287, 172–181. [Google Scholar] [CrossRef] [PubMed]
  44. Zuo, T.; Wang, L.; Morrison, C.; Chang, X.; Zhang, H.; Li, W.; Liu, Y.; Wang, Y.; Liu, X.; Chan, M.W.Y.; et al. FOXP3 Is an X-Linked Breast Cancer Suppressor Gene and an Important Repressor of the HER-2/ErbB2 Oncogene. Cell 2007, 129, 1275–1286. [Google Scholar] [CrossRef] [Green Version]
  45. Dolinsek, T.; Markelc, B.; Bosnjak, M.; Blagus, T.; Prosen, L.; Kranjc, S.; Stimac, M.; Lampreht, U.; Sersa, G.; Cemazar, M. Endoglin Silencing has Significant Antitumor Effect on Murine Mammary Adenocarcinoma Mediated by Vascular Targeted Effect. Curr. Gene Ther. 2015, 15, 228–244. [Google Scholar] [CrossRef]
  46. Lucá, R.; Averna, M.; Zalfa, F.; Vecchi, M.; Bianchi, F.; La Fata, G.; Del Nonno, F.; Nardacci, R.; Bianchi, M.; Nuciforo, P.; et al. The Fragile X Protein binds mRNAs involved in cancer progression and modulates metastasis formation. EMBO Mol. Med. 2013, 5, 1523–1536. [Google Scholar] [CrossRef] [Green Version]
  47. Cottone, L.; Capobianco, A.; Gualteroni, C.; Monno, A.; Raccagni, I.; Valtorta, S.; Canu, T.; Di Tomaso, T.; Lombardo, A.; Esposito, A.; et al. Leukocytes recruited by tumor-derived HMGB1 sustain peritoneal carcinomatosis. Oncoimmunology 2016, 5, e1122860. [Google Scholar] [CrossRef] [Green Version]
  48. Li, W.; Wang, L.; Katoh, H.; Liu, R.; Zheng, P.; Liu, Y. Identification of a tumor suppressor relay between the FOXP3 and the Hippo pathways in breast and prostate cancers. Cancer Res. 2011, 71, 2162–2171. [Google Scholar] [CrossRef]
  49. Dalziel, M.; Yeahuang, R.; Dall’Olio, F.; Morris, J.R.; Taylor-Papadimitriou, J.; Lau, J.T.Y. Mouse ST6Gal sialyltransferase gene expression during mammary gland lactation. Glycobiology 2001, 11, 407–412. [Google Scholar] [CrossRef] [Green Version]
  50. Znidar, K.; Bosnjak, M.; Semenova, N.; Pakhomova, O.; Heller, L.; Cemazar, M. Tumor cell death after electrotransfer of plasmid DNA is associated with cytosolic DNA sensor upregulation. Oncotarget 2018, 9, 18665–18681. [Google Scholar] [CrossRef] [Green Version]
  51. Servier Medical Art. Licensed under a Creative Commons Attribution 3.0 Unported License. Available online: http://smart.servier.com (accessed on 24 October 2019).
  52. Schirmbeck, R.; Riedl, P.; Kupferschmitt, M.; Wegenka, U.; Hauser, H.; Rice, J.; Kröger, A.; Reimann, J. Priming Protective CD8 T Cell Immunity by DNA Vaccines Encoding Chimeric, Stress Protein-Capturing Tumor-Associated Antigen. J. Immunol. 2006, 177, 1534–1542. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Serafini, P.; De Santo, C.; Marigo, I.; Cingarlini, S.; Dolcetti, L.; Gallina, G.; Zanovello, P.; Bronte, V. Derangement of immune responses by myeloid suppressor cells. Cancer Immunol. Immunother. 2004, 53, 64–72. [Google Scholar] [CrossRef] [PubMed]
  54. Shibue, T.; Brooks, M.W.; Fatih Inan, M.; Reinhardt, F.; Weinberg, R.A. The outgrowth of micrometastases is enabled by the formation of filopodium-like protrusions. Cancer Discov. 2012, 2, 706–721. [Google Scholar] [CrossRef]
  55. Movahedi, K.; Laoui, D.; Gysemans, C.; Baeten, M.; Stangé, G.; Van Den Bossche, J.; Mack, M.; Pipeleers, D.; In’t Veld, P.; De Baetselier, P.; et al. Different tumor microenvironments contain functionally distinct subsets of macrophages derived from Ly6C(high) monocytes. Cancer Res. 2010, 70, 5728–5739. [Google Scholar] [CrossRef] [PubMed]
  56. Galdiero, M.R.; Bonavita, E.; Barajon, I.; Garlanda, C.; Mantovani, A.; Jaillon, S. Tumor associated macrophages and neutrophils in cancer. Immunobiology 2013, 218, 1402–1410. [Google Scholar] [CrossRef]
  57. Bronte, V.; Brandau, S.; Chen, S.H.; Colombo, M.P.; Frey, A.B.; Greten, T.F.; Mandruzzato, S.; Murray, P.J.; Ochoa, A.; Ostrand-Rosenberg, S.; et al. Recommendations for myeloid-derived suppressor cell nomenclature and characterization standards. Nat. Commun. 2016, 7, 12150. [Google Scholar] [CrossRef] [Green Version]
  58. Schmieder, A.; Schledzewski, K.; Michel, J.; Tuckermann, J.P.; Tome, L.; Sticht, C.; Gkaniatsou, C.; Nicolay, J.P.; Demory, A.; Faulhaber, J.; et al. Synergistic activation by p38MAPK and glucocorticoid signaling mediates induction of M2-like tumor-associated macrophages expressing the novel CD20 homolog MS4A8A. Int. J. Cancer 2011, 129, 122–132. [Google Scholar] [CrossRef]
  59. Guiducci, C.; Vicari, A.P.; Sangaletti, S.; Trinchieri, G.; Colombo, M.P. Redirecting in vivo elicited tumor infiltrating macrophages and dendritic cells towards tumor rejection. Cancer Res. 2005, 65, 3437–3446. [Google Scholar] [CrossRef] [Green Version]
  60. Fuchs, T.; Hahn, M.; Riabov, V.; Yin, S.; Kzhyshkowska, J.; Busch, S.; Püllmann, K.; Beham, A.W.; Neumaier, M.; Kaminski, W.E. A combinatorial αβ T cell receptor expressed by macrophages in the tumor microenvironment. Immunobiology 2017, 222, 39–44. [Google Scholar] [CrossRef]
  61. Riabov, V.; Yin, S.; Song, B.; Avdic, A.; Schledzewski, K.; Ovsiy, I.; Gratchev, A.; Verdiell, M.L.; Sticht, C.; Schmuttermaier, C.; et al. Stabilin-1 is expressed in human breast cancer and supports tumor growth in mammary adenocarcinoma mouse model. Oncotarget 2016, 7, 31097–31110. [Google Scholar] [CrossRef] [Green Version]
  62. Liu, C.; Yu, S.; Kappes, J.; Wang, J.; Grizzle, W.E.; Zinn, K.R.; Zhang, H.G. Expansion of spleen myeloid suppressor cells represses NK cell cytotoxicity in tumor-bearing host. Blood 2007, 109, 4336–4342. [Google Scholar] [CrossRef] [PubMed]
  63. Bauer, R.; Udonta, F.; Wroblewski, M.; Ben-Batalla, I.; Santos, I.M.; Taverna, F.; Kuhlencord, M.; Gensch, V.; Päsler, S.; Vinckier, S.; et al. Blockade of myeloid-derived suppressor cell expansion with all-trans retinoic acid increases the efficacy of antiangiogenic therapy. Cancer Res. 2018, 78, 3220–3232. [Google Scholar] [PubMed] [Green Version]
  64. Rong, L.; Bian, Y.; Liu, S.; Liu, X.; Li, X.; Liu, H.; Zhou, J.; Peng, J.; Zhang, H.; Chen, H.; et al. Identifying tumor promoting genomic alterations in tumorassociated fibroblasts via retrovirus-insertional mutagenesis. Oncotarget 2017, 8, 97231–97245. [Google Scholar] [CrossRef] [Green Version]
  65. Wang, Y.Y.; Attané, C.; Milhas, D.; Dirat, B.; Dauvillier, S.; Guerard, A.; Gilhodes, J.; Lazar, I.; Alet, N.; Laurent, V.; et al. Mammary adipocytes stimulate breast cancer invasion through metabolic remodeling of tumor cells. JCI Insight 2017, 2, e87489. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Wolfers, J.; Lozier, A.; Raposo, G.; Regnault, A.; Théry, C.; Masurier, C.; Flament, C.; Pouzieux, S.; Faure, F.; Tursz, T.; et al. Tumor-derived exosomes are a source of shared tumor rejection antigens for CTL cross-priming. Nat. Med. 2001, 7, 297–303. [Google Scholar] [CrossRef] [PubMed]
  67. Liu, C.; Yu, S.; Zinn, K.; Wang, J.; Zhang, L.; Jia, Y.; Kappes, J.C.; Barnes, S.; Kimberly, R.P.; Grizzle, W.E.; et al. Murine Mammary Carcinoma Exosomes Promote Tumor Growth by Suppression of NK Cell Function. J. Immunol. 2006, 176, 1375–1385. [Google Scholar] [CrossRef] [Green Version]
  68. Yu, S.; Liu, C.; Su, K.; Wang, J.; Liu, Y.; Zhang, L.; Li, C.; Cong, Y.; Kimberly, R.; Grizzle, W.E.; et al. Tumor Exosomes Inhibit Differentiation of Bone Marrow Dendritic Cells. J. Immunol. 2007, 178, 6867–6875. [Google Scholar] [CrossRef] [Green Version]
  69. Diamond, J.M.; Vanpouille-Box, C.; Spada, S.; Rudqvist, N.P.; Chapman, J.R.; Ueberheide, B.M.; Pilones, K.A.; Sarfraz, Y.; Formenti, S.C.; Demaria, S. Exosomes Shuttle TREX1-Sensitive IFN-Stimulatory dsDNA from Irradiated Cancer Cells to DCs. Cancer Immunol. Res. 2018, 6, 910–920. [Google Scholar] [CrossRef] [Green Version]
  70. Giovarelli, M.; Cappello, P.; Forni, G.; Salcedo, T.; Moore, P.A.; LeFleur, D.W.; Nardelli, B.; Di Carlo, E.; Lollini, P.-L.; Ruben, S.; et al. Tumor Rejection and Immune Memory Elicited by Locally Released LEC Chemokine Are Associated with an Impressive Recruitment of APCs, Lymphocytes, and Granulocytes. J. Immunol. 2000, 164, 3200–3206. [Google Scholar] [CrossRef] [Green Version]
  71. Cappello, P.; Caorsi, C.; Bosticardo, M.; De Angelis, S.; Novelli, F.; Forni, G.; Giovarelli, M. CCL16/LEC powerfully triggers effector and antigen-presenting functions of macrophages and enhances T cell cytotoxicity. J. Leukoc. Biol. 2004, 75, 135–142. [Google Scholar] [CrossRef] [Green Version]
  72. Guiducci, C.; Di Carlo, E.; Parenza, M.; Hitt, M.; Giovarelli, M.; Musiani, P.; Colombo, M.P. Intralesional Injection of Adenovirus Encoding CC Chemokine Ligand 16 Inhibits Mammary Tumor Growth and Prevents Metastatic-Induced Death after Surgical Removal of the Treated Primary Tumor. J. Immunol. 2004, 172, 4026–4036. [Google Scholar] [CrossRef] [PubMed]
  73. Musiani, P.; Modesti, A.; Brunetti, M.; Modica, A.; Vitullo, P.; Gulino, A.; Bosco, M.C.; Colombo, M.P.; Nanni, P.; Cavallo, F.; et al. Nature and potential of the reactive response to mouse mammary adenocarcinoma cells engineered with interleukin-2, interleukin-4 or interferon-γ genes. Nat. Immun. 1994, 13, 93–101. [Google Scholar] [PubMed]
  74. Allione, A.; Consalvo, M.; Nanni, P.; Lollini, P.L.; Cavallo, F.; Giovarelli, M.; Forni, M.; Gulino, A.; Colombo, M.P.; Dellabona, P.; et al. Immunizing and Curative Potential of Replicating and Nonreplicating Murine Mammary Adenocarcinoma Cells Engineered with Interleukin (IL)-2, IL-4, IL-6, IL-7, IL-10, Tumor Necrosis Factor a, Granulocyte-Macrophage Colony-stimulating Factor, and y-Interfero. Cancer Res. 1994, 54, 6022–6026. [Google Scholar] [PubMed]
  75. Pericle, F.; Kirken, R.A.; Epling-Burnette, P.K.; Blanchard, D.K.; Djeu, J.Y. Direct killing of interleukin-2-transfected tumor cells by human neutrophils. Int. J. Cancer 1996, 66, 367–373. [Google Scholar] [CrossRef]
  76. Vagliani, M.; Rodolfo, M.; Cavallo, F.; Parenza, M.; Melani, C.; Parmiani, G.; Forni, G.; Colombo, M.P. Interleukin 12 potentiates the curative effect of a vaccine based on interleukin 2-transduced tumor cells. Cancer Res. 1996, 56, 467–470. [Google Scholar]
  77. Provinciali, M.; Argentati, K.; Tibaldi, A. Efficacy of cancer gene therapy in aging adenocarcinoma cells engineered to release IL-2 are rejected but do not induce tumor specific immune memory in old mice. Gene Ther. 2000, 7, 624–632. [Google Scholar] [CrossRef] [Green Version]
  78. Modesti, A.; D’Orazi, G.; Masuelli, L.; Modica, A.; Scarpa, S.; Bosco, M.C.; Forni, G. Ultrastructural evidence of the mechanisms responsible for interleukin-4-activated rejection of a spontaneous murine adenocarcinoma. Int. J. Cancer 1993, 53, 988–993. [Google Scholar] [CrossRef]
  79. Pericle, F.; Giovarelli, M.; Cavallo, F.; Di Pierro, F.; Novelli, F.; Forni, G.; Colombo, M.P.; Ferrari, G.; Musiani, P.; Modesti, A. An efficient Th2-type memory follows CD8+lymphocyte-driven and eosinophil-mediated rejection of a spontaneous mouse mammary adenocarcinoma engineered to release IL-4. J. Immunol. 1994, 153, 5659–5673. [Google Scholar]
  80. Musiani, P.; Allione, A.; Modica, A.; Lollini, P.L.; Giovarelli, M.; Cavallo, F.; Belardelli, F.; Forni, G.; Modesti, A. Role of Neutrophils and Lymphocytes in Inhibition of a Mouse Mammary Adenocarcinoma Engineered to Release IL-2, IL-4, IL-7, IL-10, IFN-α, IFN-γ, and TNF-α. Lab. Investig. 1996, 74, 146–157. [Google Scholar]
  81. Cayeux, S.; Richter, G.; Noffz, G.; Dörken, B.; Blankenstein, T. Influence of gene-modified (IL-7, IL-4, and B7) tumor cell vaccines on tumor antigen presentation. J. Immunol. 1997, 158, 2834–2841. [Google Scholar]
  82. Belardelli, F.; Ferrantini, M.; Santini, S.M.; Baccarini, S.; Proietti, E.; Colombo, M.P.; Sprent, J.; Tough, D.F. The induction of in vivo proliferation of long-lived CD44(hi) CD8+ T cells after the injection of tumor cells expressing IFN-α1 into syngeneic mice. Cancer Res. 1998, 58, 5795–5802. [Google Scholar] [PubMed]
  83. Di Carlo, E.; Modesti, A.; Coletti, A.; Colombo, M.P.; Giovarelli, M.; Forni, G.; Diodoro, M.G.; Musiani, P. Interaction between endothelial cells and the secreted cytokine drives the fate of an IL4- or an IL5-transduced tumour. J. Pathol. 1998, 186, 390–397. [Google Scholar] [CrossRef]
  84. Pacor, S.; Gagliardi, R.; Di Daniel, E.; Vadori, M.; Sava, G. In vitro down regulation of ICAM-1 and E-cadherin and in vivo reduction of lung metastases of TS/A adenocarcinoma by a lysozyme derivative. Int. J. Mol. Med. 1999, 4, 369–375. [Google Scholar] [CrossRef] [PubMed]
  85. Pacor, S.; Magnarin, M.; Carotenuto, M.E.; Spessotto, P.; Zabucchi, G.; Sava, G. In vitro growth of TS/A adenocarcinoma and of the gene transfected TS/A-IL4 line on biological substrates. Anticancer Res. 2000, 20, 191–196. [Google Scholar]
  86. Krüger-Krasagakes, S.; Li, W.; Richter, G.; Diamantstein, T.; Blankenstein, T. Eosinophils infiltrating interleukin-5 gene-transfected tumors do not suppress tumor growth. Eur. J. Immunol. 1993, 23, 992–995. [Google Scholar] [CrossRef]
  87. Di Carlo, E.; Coletti, A.; Modesti, A.; Giovarelli, M.; Forni, G.; Musiani, P. Local release of interleukin-10 by transfected mouse adenocarcinoma cells exhibits pro- and anti-inflammatory activity and results in a delayed tumor rejection. Eur. Cytokine Netw. 1998, 9, 61–68. [Google Scholar]
  88. Hock, H.; Dorsch, M.; Diamantstein, T.; Blankenstein, T. Interleukin 7 induces CD4 + T cell-dependent tumor rejection. J. Exp. Med. 1991, 174, 1291–1298. [Google Scholar] [CrossRef] [Green Version]
  89. Cayeux, S.; Beck, C.; Aicher, A.; Dörken, B.; Blankenstein, T. Tumor cells cotransfected with interleukin-7 and B7.1 genes induce CD25 and CD28 on tumor-infiltrating T lymphocytes and are strong vaccines. Eur. J. Immunol. 1995, 25, 2325–2331. [Google Scholar] [CrossRef]
  90. Willimsky, G.; Blankenstein, T. Interleukin-7/B7.1-encoding adenoviruses induce rejection of transplanted but not nontransplanted tumors. Cancer Res. 2000, 60, 685–692. [Google Scholar]
  91. Giovarelli, M.; Musiani, P.; Modesti, A.; Dellabona, P.; Casorati, G.; Allione, A.; Consalvo, M.; Cavallo, F.; di Pierro, F.; De Giovanni, C. Local release of IL-10 by transfected mouse mammary adenocarcinoma cells does not suppress but enhances antitumor reaction and elicits a strong cytotoxic lymphocyte and antibody-dependent immune memory. J. Immunol. 1995, 155, 3112–3123. [Google Scholar]
  92. Morini, M.; Albini, A.; Lorusso, G.; Moelling, K.; Lu, B.; Cilli, M.; Ferrini, S.; Noonan, D.M. Prevention of angiogenesis by naked DNA IL-12 gene transfer: Angioprevention by immunogene therapy. Gene Ther. 2004, 11, 284–291. [Google Scholar] [CrossRef] [Green Version]
  93. Weber, S.M.; Qi, C.; Neal, Z.; Sondel, P.; Mahvi, D.M. IL-12 cDNA direct injection: Antimetastatic effect from a single injection in a murine hepatic metastases model. J. Surg. Res. 2004, 122, 210–217. [Google Scholar] [CrossRef]
  94. Oshikawa, K.; Shi, F.; Rakhmilevich, A.L.; Sondel, P.M.; Mahvi, D.M.; Yang, N.S. Synergistic inhibition of tumor growth in a murine mammary adenocarcinoma model by combinational gene therapy using IL-12, pro-IL-18, and IL-1β converting enzyme cDNA. Proc. Natl. Acad. Sci. USA 1999, 96, 13351–13356. [Google Scholar] [CrossRef] [Green Version]
  95. Comes, A.; Di Carlo, E.; Musiani, P.; Rosso, O.; Meazza, R.; Chiodoni, C.; Colombo, M.P.; Ferrini, S. IFN-γ-independent synergistic effects of IL-12 and IL-15 induce anti-tumor immune responses in syngeneic mice. Eur. J. Immunol. 2002, 32, 1914–1923. [Google Scholar] [CrossRef]
  96. Cavallo, F.; Signorelli, P.; Giovarelli, M.; Musiani, P.; Modesti, A.; Brunda, M.J.; Colombo, M.P.; Forni, G. Antitumor efficacy of adenocarcinoma cells engineered to produce interleukin 12 (IL-12) or other cytokines compared with exogenous IL-12. J. Natl. Cancer Inst. 1997, 89, 1049–1058. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Gri, G.; Chiodoni, C.; Gallo, E.; Stoppacciaro, A.; Liew, F.Y.; Colombo, M.P. Antitumor effect of interleukin (IL)-12 in the absence of endogenous IFN-γ: A role for intrinsic tumor immunogenicity and IL-15. Cancer Res. 2002, 62, 4390–4397. [Google Scholar]
  98. Puisieux, I.; Odin, L.; Poujol, D.; Moingeon, P.; Tartaglia, J.; Cox, W.; Favrot, M. Canarypox virus-mediated interleukin 12 gene transfer into murine mammary adenocarcinoma induces tumor suppression and long-term antitumoral immunity. Hum. Gene Ther. 1998, 9, 2481–2492. [Google Scholar] [CrossRef]
  99. De Giovanni, C.; Nicoletti, G.; Landuzzi, L.; Rossi, I.; Astolfi, A.; Ricci, C.; Di Carlo, E.; Musiani, P.; Forni, G.; Fradelizi, D.; et al. Therapy of lung metastases through combined vaccination with carcinoma cells engineered to release IL-13 and IFN-γ. Gene Ther. 2001, 8, 1698–1704. [Google Scholar] [CrossRef]
  100. Morris, J.C.; Ramlogan-Steel, C.A.; Yu, P.; Black, B.A.; Mannan, P.; Allison, J.P.; Waldmann, T.A.; Steel, J.C. Vaccination with tumor cells expressing IL-15 and IL-15R inhibits murine breast and prostate cancer. Gene Ther. 2014, 21, 393–401. [Google Scholar] [CrossRef]
  101. Meazza, R.; Lollini, P.L.; Nanni, P.; De Giovanni, C.; Gaggero, A.; Comes, A.; Cilli, M.; Di Carlo, E.; Ferrini, S.; Musiani, P. Gene transfer of a secretable form of IL-15 in murine adenocarcinoma cells: Effects on tumorigenicity, metastatic potential and immune response. Int. J. Cancer 2000, 87, 574–581. [Google Scholar] [CrossRef]
  102. Di Carlo, E.; Comes, A.; Orengo, A.M.; Rosso, O.; Meazza, R.; Musiani, P.; Colombo, M.P.; Ferrini, S. IL-21 Induces Tumor Rejection by Specific CTL and IFN-γ-Dependent CXC Chemokines in Syngeneic Mice. J. Immunol. 2004, 172, 1540–1547. [Google Scholar] [CrossRef] [PubMed]
  103. Ferrantini, M.; Giovarelli, M.; Modesti, A.; Musiani, P.; Modica, A.; Venditti, M.; Peretti, E.; Lollini, P.L.; Nanni, P.; Forni, G.; et al. IFN-α1 gene expression into a metastatic murine adenocarcinoma (TS/A) results in CD8+T cell-mediated tumor rejection and development of antitumor immunity: Comparative studies with IFN-γ-producing TS/A cells. J. Immunol. 1994, 153, 4604–4615. [Google Scholar]
  104. Santodonato, L.; D’Agostino, G.; Santini, S.M.; Carlei, D.; Musiani, P.; Modesti, A.; Signorelli, P.; Belardelli, F.; Ferrantini, M. Local and systemic antitumor response after combined therapy of mouse metastatic tumors with tumor cells expressing IFN-α and HSVtk: Perspectives for the generation of cancer vaccines. Gene Ther. 1997, 4, 1246–1255. [Google Scholar] [CrossRef] [Green Version]
  105. Scarpa, S.; Giuffrida, A.; Palumbo, C.; Vasaturo, F.; Signorelli, P.; Forni, G.; Modesti, M.; Ferrantini, M.; Belardelli, F.; Musiani, P.; et al. Extracellular matrix remodelling in a murine mammary adenocarcinoma transfected with the interferon-alpha1 gene. J. Pathol. 1997, 181, 116–123. [Google Scholar] [CrossRef]
  106. Rossi, I.; Nicoletti, G.; Landuzzi, L.; Frabetti, F.; De Giovanni, C.; Nanni, P.; Musiani, P.; Ferrantini, M.; Belardelli, F.; Lollini, P.L. Inhibition of lung colonisation of a mouse mammary carcinoma by therapeutic vaccination with interferon-α gene-transduced tumor cells. Clin. Exp. Metastasis 1998, 16, 123–128. [Google Scholar] [CrossRef]
  107. Tüting, T.; Gambotto, A.; Baar, J.; Davis, I.D.; Storkus, W.J.; Zavodny, P.J.; Narula, S.; Tahara, H.; Robbins, P.D.; Lotze, M.T. Interferon-α gene therapy for cancer: Retroviral transduction of fibroblasts and particle-mediated transfection of tumor cells are both effective strategies for gene delivery in murine tumor models. Gene Ther. 1997, 4, 1053–1060. [Google Scholar] [CrossRef] [Green Version]
  108. Coleman, M.; Muller, S.; Quezada, A.; Mendiratta, S.K.; Wang, J.; Thull, N.M.; Bishop, J.; Matar, M.; Mester, J.; Pericle, F. Nonviral interferon α gene therapy inhibits growth of established tumors by eliciting a systemic immune response. Hum. Gene Ther. 1998, 9, 2223–2230. [Google Scholar] [CrossRef]
  109. Rozera, C.; Carlei, D.; Lollini, P.L.; De Giovanni, C.; Musiani, P.; Di Carlo, E.; Belardelli, F.; Ferrantini, M. Interferon (IFN)-β gene transfer into TS/A adenocarcinoma cells and comparison with IFN-α. Differential effects on tumorigenicity and host response. Am. J. Pathol. 1999, 154, 1211–1222. [Google Scholar] [CrossRef]
  110. Lollini, P.L.; Nanni, P. Minimal requirements for characterization of cytokine gene-transduced tumor cells: A proposal. J. Natl. Cancer Inst. 1995, 87, 1717–1718. [Google Scholar] [CrossRef]
  111. Nanni, P.; De Giovanni, C.; Landuzzi, L.; Nicoletti, G.; Frabetti, F.; Rossi, I.; Cavallo, F.; Giovarelli, M.; Forni, G.; Lollini, P.L. Therapy of murine mammary carcinoma metastasis with interferon γ and MHC gene-transduced tumour cells. Br. J. Cancer 1996, 74, 1564–1569. [Google Scholar] [CrossRef] [Green Version]
  112. Nanni, P.; De Giovanni, C.; Nicoletti, G.; Landuzzi, L.; Rossi, I.; Frabetti, F.; Giovarelli, M.; Forni, G.; Cavallo, F.; Di Carlo, E.; et al. The immune response elicited by mammary adenocarcinoma cells transduced with interferon-γ and cytosine deaminase genes cures lung metastases by parental cells. Hum. Gene Ther. 1998, 9, 217–224. [Google Scholar] [CrossRef] [PubMed]
  113. Sacchi, A.; Gasparri, A.; Curnis, F.; Bellone, M.; Corti, A. Crucial role for interferon γ in the synergism between tumor vasculature-targeted tumor necrosis factor α (NGR-TNF) and doxorubicin. Cancer Res. 2004, 64, 7150–7155. [Google Scholar] [CrossRef]
  114. Cavallo, F.; Martin-Fontecha, A.; Bellone, M.; Heltai, S.; Gatti, E.; Tornaghi, P.; Freschi, M.; Forni, G.; Dellabona, P.; Casorati, G. Co-expression of B7-1 and ICAM-1 on tumors is required for rejection and the establishment of a memory response. Eur. J. Immunol. 1995, 25, 1154–1162. [Google Scholar] [CrossRef]
  115. Martin-Fontecha, A.; Cavallo, F.; Bellone, M.; Heltai, S.; Lezzi, G.; Tornaghi, P.; Nabavi, N.; Forni, G.; Dellabona, P.; Casorati, G. Heterogeneous effects of B7-1 and B7-2 in the induction of both protective and therapeutic antitumor immunity against different mouse tumors. Eur. J. Immunol. 1996, 26, 1851–1859. [Google Scholar] [CrossRef] [PubMed]
  116. Martín-Fontecha, A.; Moro, M.; Crosti, M.C.; Veglia, F.; Casorati, G.; Dellabona, P. Vaccination with Mouse Mammary Adenocarcinoma Cells Coexpressing B7-1 (CD80) and B7-2 (CD86) Discloses the Dominant Effect of B7-1 in the Induction of Antitumor Immunity. J. Immunol. 2000, 164, 698–704. [Google Scholar] [CrossRef] [Green Version]
  117. Lollini, P.L.; De Giovanni, C.; Landuzzi, L.; Nicoletti, G.; Frabetti, F.; Nanni, P.; Landuzzi, L.; Nicoletti, G.; Cavallo, F.; Giovarelli, M.; et al. Transduction of Genes Coding for a Histocompatibility (MHC) Antigen and for Its Physiological Inducer Interferon-γ in the Same Cell: Efficient MHC Expression and Inhibition of Tumor and Metastasis Growth. Hum. Gene Ther. 1995, 6, 743–752. [Google Scholar] [CrossRef]
  118. Couderc, B.; Zitvogel, L.; Douin-Echinard, V.; Djennane, L.; Tahara, H.; Favre, G.; Lotze, M.T.; Robbins, P.D. Enhancement of antitumor immunity by expression of CD70 (CD27 ligand) or CD154 (CD40 ligand) costimulatory molecules in tumor cells. Cancer Gene Ther. 1998, 5, 163–175. [Google Scholar]
  119. Douin-Echinard, V.; Bornes, S.; Rochaix, P.; Tilkin, A.F.; Peron, J.M.; Bonnet, J.; Favre, G.; Couderc, B. The expression of CD70 and CD80 by gene-modified tumor cells induces an antitumor response depending on the MHC status. Cancer Gene Ther. 2000, 7, 1543–1556. [Google Scholar] [CrossRef] [Green Version]
  120. Grangeon, C.; Cormary, C.; Douin-Echinard, V.; Favre, G.; Couderc, B.; Tilkin-Mariamé, A.F. In vivo induction of antitumor immunity and protection against tumor growth by injection of CD154-expressing tumor cells. Cancer Gene Ther. 2002, 9, 282–288. [Google Scholar] [CrossRef] [Green Version]
  121. Giovarelli, M.; Cappello, P.; Rigamonti, L.; Bernabei, P.; Novelli, F.; Lollini, P.L.; Forni, G.; Musiani, P.; Di Carlo, E.; Garotta, G.; et al. A “stealth effect”: Adenocarcinoma cells engineered to express TRAIL elude tumor-specific and allogeneic T cell reactions. J. Immunol. 1999, 163, 4886–4893. [Google Scholar]
  122. Prigent, P.; El Mir, S.; Dréano, M.; Triebel, F. Lymphocyte activation gene-3 induces tumor regression and antitumor immune responses. Eur. J. Immunol. 1999, 29, 3867–3876. [Google Scholar] [CrossRef]
  123. Di Carlo, E.; Cappello, P.; Sorrentino, C.; D’Antuono, T.; Pellicciotta, A.; Giovarelli, M.; Forni, G.; Musiani, P.; Triebel, F. Immunological mechanisms elicited at the tumour site by lymphocyte activation gene-3 (LAG-3) versus IL-12: Sharing a common Th1 anti-tumour immune pathway. J. Pathol. 2005, 205, 82–91. [Google Scholar] [CrossRef]
  124. Croci, S.; Nicoletti, G.; Landuzzi, L.; Palladini, A.; Chiarini, F.; Nanni, P.; Lollini, P.L.; De Giovanni, C. Expression of a functional CCR7 chemokine receptor inhibits the post-intravasation steps of metastasis in malignant murine mammary cancer cells. Oncol. Rep. 2007, 18, 451–456. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Consalvo, M.; Mullen, C.A.; Modesti, A.; Musiani, P.; Allione, A.; Cavallo, F.; Giovarelli, M.; Forni, G. 5-Fluorocytosine-Induced Eradication of Murine Adenocarcinomas Engineered To Express the Cytosine Deaminase Suicide Gene Requires Host Immune Competence and Leaves an Efficient Memory. J. Immunol. 1995, 154, 5302–5312. [Google Scholar]
  126. Uckert, W.; Kammertöns, T.; Haack, K.; Qin, Z.; Gebert, J.; Schendel, D.J.; Blankenstein, T. Double suicide gene (cytosine deaminase and herpes simplex virus thymidine kinase) but not single gene transfer allows reliable elimination of tumor cells in vivo. Hum. Gene Ther. 1998, 9, 855–865. [Google Scholar] [CrossRef]
  127. Porosnicu, M.; Mian, A.; Barber, G.N. The Oncolytic Effect of Recombinant Vesicular Stomatitis Virus Is Enhanced by Expression of the Fusion Cytosine Deaminase/Uracil Phosphoribosyltransferase Suicide Gene. Cancer Res. 2003, 63, 8366–8376. [Google Scholar]
  128. Leveille, S.; Goulet, M.-L.; Lichty, B.D.; Hiscott, J. Vesicular Stomatitis Virus Oncolytic Treatment Interferes with Tumor-Associated Dendritic Cell Functions and Abrogates Tumor Antigen Presentation. J. Virol. 2011, 85, 12160–12169. [Google Scholar] [CrossRef] [Green Version]
  129. Chen, X.; Zhang, D.; Dennert, G.; Hung, G.; Lee, A.S. Eradication of murine mammary adenocarcinoma through HSVtk expression directed by the glucose-starvation inducible grp78 promoter. Breast Cancer Res. Treat. 2000, 59, 81–90. [Google Scholar] [CrossRef]
  130. Faneca, H.; Cabrita, A.S.; Simões, S.; Pedroso de Lima, M.C. Evaluation of the antitumoral effect mediated by IL-12 and HSV-tk genes when delivered by a novel lipid-based system. Biochim. Biophys. Acta Biomembr. 2007, 1768, 1093–1102. [Google Scholar] [CrossRef] [Green Version]
  131. Meazza, R.; Comes, A.; Orengo, A.M.; Ferrini, S.; Accolla, R.S. Tumor rejection by gene transfer of the MHC class II transactivator in murine mammary adenocarcinoma cells. Eur. J. Immunol. 2003, 33, 1183–1192. [Google Scholar] [CrossRef]
  132. Mortara, L.; Frangione, V.; Castellani, P.; De Lerma Barbaro, A.; Accolla, R.S. Irradiated CIITA-positive mammary adenocarcinoma cells act as a potent anti-tumor-preventive vaccine by inducing tumor-specific CD4+ T cell priming and CD8+ T cell effector functions. Int. Immunol. 2009, 21, 655–665. [Google Scholar] [CrossRef] [Green Version]
  133. McCaw, T.R.; Li, M.; Starenki, D.; Cooper, S.J.; Liu, M.; Meza-Perez, S.; Arend, R.C.; Buchsbaum, D.J.; Forero, A.; Randall, T.D. The expression of MHC class II molecules on murine breast tumors delays T-cell exhaustion, expands the T-cell repertoire, and slows tumor growth. Cancer Immunol. Immunother. 2019, 68, 175–188. [Google Scholar] [CrossRef] [PubMed]
  134. Lipnik, K.; Naschberger, E.; Gonin-Laurent, N.; Kodajova, P.; Petznek, H.; Rungaldier, S.; Astigiano, S.; Ferrini, S.; Stürz, M.; Hohenadl, C. Interferon γ-Induced human guanylate binding protein 1 inhibits mammary tumor growth in mice. Mol. Med. 2010, 16, 177–187. [Google Scholar] [CrossRef] [PubMed]
  135. Kim, P.K.M.; Armstrong, M.; Liu, Y.; Yan, P.; Bucher, B.; Zuckerbraun, B.S.; Gambotto, A.; Billiar, T.R.; Yim, J.H. IRF-1 expression induces apoptosis and inhibits tumor growth in mouse mammary cancer cells in vitro and in vivo. Oncogene 2004, 23, 1125–1135. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  136. Lollini, P.-L.; D’Errico, A.; De Giovanni, C.; Landuzzi, L.; Frabetti, F.; Nicoletti, G.; Cavallo, F.; Giovarelli, M.; Grigioni, W.F.; Nanni, P. Systemic effects of cytokines released by gene-transduced tumor cells: Marked hyperplasia induced in small bowel by γ-interferon transfectants through host lymphocytes. Int. J. Cancer 1995, 61, 425–430. [Google Scholar] [CrossRef]
  137. Musiani, P.; Modesti, A.; Giovarelli, M.; Cavallo, F.; Forni, G.; Lollini, P.L.; Colombo, M.P. Cytokines, tumour-cell death and immunogenicity: A question of choice. Immunol. Today 1997, 18, 32–36. [Google Scholar] [CrossRef]
  138. Dranoff, G.; Jaffee, E.; Lazenby, A.; Golumbek, P.; Levitsky, H.; Brose, K.; Jackson, V.; Hamada, H.; Pardoll, D.; Mulligan, R.C. Vaccination with irradiated tumor cells engineered to secrete murine granulocyte-macrophage colony-stimulating factor stimulates potent, specific, and long-lasting anti-tumor immunity. Proc. Natl. Acad. Sci. USA 1993, 90, 3539–3543. [Google Scholar] [CrossRef] [Green Version]
  139. Heiber, J.F.; Barber, G.N. Vesicular Stomatitis Virus Expressing Tumor Suppressor p53 Is a Highly Attenuated, Potent Oncolytic Agent. J. Virol. 2011, 85, 10440–10450. [Google Scholar] [CrossRef] [Green Version]
  140. Bonnet, M.E.; Gossart, J.B.; Benoit, E.; Messmer, M.; Zounib, O.; Moreau, V.; Behr, J.P.; Lenne-Samuel, N.; Kedinger, V.; Meulle, A.; et al. Systemic delivery of sticky siRNAs targeting the cell cycle for lung tumor metastasis inhibition. J. Control. Release 2013, 170, 183–190. [Google Scholar] [CrossRef]
  141. Kéramidas, M.; De Fraipont, F.; Karageorgis, A.; Moisan, A.; Persoons, V.; Richard, M.J.; Coll, J.L.; Rome, C. The dual effect of mscs on tumour growth and tumour angiogenesis. Stem Cell Res. Ther. 2013, 4, 41. [Google Scholar] [CrossRef] [Green Version]
  142. Josserand, V.; Texier-Nogues, I.; Huber, P.; Favrot, M.C.; Coll, J.L. Non-invasive in vivo optical imaging of the lacZ and luc gene expression in mice. Gene Ther. 2007, 14, 1587–1593. [Google Scholar] [CrossRef] [PubMed]
  143. Steel, J.C.; Morrison, B.J.; Mannan, P.; Abu-Asab, M.S.; Wildner, O.; Miles, B.K.; Yim, K.C.; Ramanan, V.; Prince, G.A.; Morris, J.C. Immunocompetent syngeneic cotton rat tumor models for the assessment of replication-competent oncolytic adenovirus. Virology 2007, 369, 131–142. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  144. Kamensek, U.; Sersa, G.; Vidic, S.; Tevz, G.; Kranjc, S.; Cemazar, M. Irradiation, cisplatin, and 5-azacytidine upregulate cytomegalovirus promoter in tumors and muscles: Implementation of non-invasive fluorescence imaging. Mol. Imaging Biol. 2011, 13, 43–52. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. Kamensek, U.; Sersa, G.; Cemazar, M. Evaluation of p21 promoter for interleukin 12 radiation induced transcriptional targeting in a mouse tumor model. Mol. Cancer 2013, 12, 136. [Google Scholar] [CrossRef] [Green Version]
  146. Bobrie, A.; Krumeich, S.; Reyal, F.; Recchi, C.; Moita, L.F.; Seabra, M.C.; Ostrowski, M.; Théry, C. Rab27a supports exosome-dependent and -independent mechanisms that modify the tumor microenvironment and can promote tumor progression. Cancer Res. 2012, 72, 4920–4930. [Google Scholar] [CrossRef]
  147. Germano, G.; Lamba, S.; Rospo, G.; Barault, L.; Magri, A.; Maione, F.; Russo, M.; Crisafulli, G.; Bartolini, A.; Lerda, G.; et al. Inactivation of DNA repair triggers neoantigen generation and impairs tumour growth. Nature 2017, 552, 1–5. [Google Scholar] [CrossRef]
  148. Cayeux, S.; Richter, G.; Becker, C.; Pezzutto, A.; Dörken, B.; Blankenstein, T. Direct and indirect T cell priming by dendritic cell vaccines. Eur. J. Immunol. 1999, 29, 225–234. [Google Scholar] [CrossRef]
  149. Cayeux, S.; Qin, Z.; Dörken, B.; Blankenstein, T. Decreased generation of anti-tumor immunity after intrasplenic immunization. Eur. J. Immunol. 2001, 31, 1392–1399. [Google Scholar] [CrossRef]
  150. Preiss, S.; Kammertoens, T.; Lampert, C.; Willimsky, G.; Blankenstein, T. Tumor-induced antibodies resemble the response to tissue damage. Int. J. Cancer 2005, 115, 456–462. [Google Scholar] [CrossRef]
  151. Dobrzanski, M.J.; Reome, J.B.; Hylind, J.C.; Rewers-Felkins, K.A. CD8-Mediated Type 1 Antitumor Responses Selectively Modulate Endogenous Differentiated and Nondifferentiated T Cell Localization, Activation, and Function in Progressive Breast Cancer. J. Immunol. 2006, 177, 8191–8201. [Google Scholar] [CrossRef]
  152. Dobrzanski, M.J.; Reome, J.B.; Hylind, J.C.; Rewers-Felkins, K.A.; Abdulsamad, K.; Adams, S.L. Ag-specific type 1 CD8 effector cells enhance methotrexate-mediated antitumor responses by modulating endogenous CD49b-expressing CD4 and CD8 T effector cell subpopulations producing IL-10. Immunol. Investig. 2008, 37, 315–338. [Google Scholar] [CrossRef] [PubMed]
  153. Benigni, F.; Zimmermann, V.S.; Hugues, S.; Caserta, S.; Basso, V.; Rivino, L.; Ingulli, E.; Malherbe, L.; Glaichenhaus, N.; Mondino, A. Phenotype and Homing of CD4 Tumor-Specific T Cells Is Modulated by Tumor Bulk. J. Immunol. 2005, 175, 739–748. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  154. Zimmermann, V.S.; Casati, A.; Schiering, C.; Caserta, S.; Hess, M.R.; Basso, V.; Mondino, A. Tumors hamper the immunogenic competence of CD4+ T cell-directed dendritic cell vaccination. J. Immunol. 2007, 179, 2899–2909. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  155. Schiering, C.; Guarnerio, J.; Basso, V.; Muzio, L.; Mondino, A. Antigen-experienced CD4+ T cells limit naïve T-cell priming in response to therapeutic vaccination in vivo. Cancer Res. 2010, 70, 6161–6170. [Google Scholar] [CrossRef] [Green Version]
  156. Martino, A.; Vismara, D.; Cicconi, R.; Delpino, A.; Ivanyi, J.; Colizzi, V.; Cassol, M.; Fraziano, M.; Piselli, P. Effective anti-tumor immunity induced in mice by a two-step vaccination protocol. In Vivo 2001, 15, 425–428. [Google Scholar]
  157. Li, Z.; Pradera, F.; Kammertoens, T.; Li, B.; Liu, S.; Qin, Z. Cross-Talk between T Cells and Innate Immune Cells Is Crucial for IFN-γ-Dependent Tumor Rejection. J. Immunol. 2007, 179, 1568–1576. [Google Scholar] [CrossRef] [Green Version]
  158. Ghiringhelli, F.; Apetoh, L.; Tesniere, A.; Aymeric, L.; Ma, Y.; Ortiz, C.; Vermaelen, K.; Panaretakis, T.; Mignot, G.; Ullrich, E.; et al. Activation of the NLRP3 inflammasome in dendritic cells induces IL-1Β-dependent adaptive immunity against tumors. Nat. Med. 2009, 15, 1170–1178. [Google Scholar] [CrossRef]
  159. Colombo, B.; Curnis, F.; Foglieni, C.; Monno, A.; Arrigoni, G.; Corti, A. Chromogranin A expression in neoplastic cells affects tumor growth and morphogenesis in mouse models. Cancer Res. 2002, 62, 941–946. [Google Scholar]
  160. Malosio, M.L.; Giordano, T.; Laslop, A.; Meldolesi, J. Dense-core granules: A specific hallmark of the neuronal/neurosecretory cell phenotype. J. Cell Sci. 2004, 117, 743–749. [Google Scholar] [CrossRef] [Green Version]
  161. Veschini, L.; Crippa, L.; Dondossola, E.; Doglioni, C.; Corti, A.; Ferrero, E. The vasostatin-1 fragment of chromogranin A preserves a quiescent phenotype in hypoxia-driven endothelial cells and regulates tumor neovascularization. FASEB J. 2011, 25, 3906–3914. [Google Scholar] [CrossRef]
  162. Melani, C.; Stoppacciaro, A.; Foroni, C.; Felicetti, F.; Caré, A.; Colombo, M.P. Angiopoietin decoy secreted at tumor site impairs tumor growth and metastases by inducing local inflammation and altering neoangiogenesis. Cancer Immunol. Immunother. 2004, 53, 600–608. [Google Scholar] [CrossRef] [PubMed]
  163. Sorli, S.C.; Le Gonidec, S.; Knibiehler, B.; Audigier, Y. Apelin is a potent activator of tumour neoangiogenesis. Oncogene 2007, 26, 7692–7699. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  164. Marionneau, S.; Ruvoën, N.; Le MoullacVaidye, B.; Clement, M.; CailleauThomas, A.; RuizPalacois, G.; Huang, P.; Jiang, X.; Le Pendu, J. Norwalk Virus binds to histo-blood group antigens present on gastroduodenal epithelial cells of secretor individuals. Gastroenterology 2002, 122, 1967–1977. [Google Scholar] [CrossRef] [PubMed]
  165. Zavaglia, D.; Favrot, M.C.; Eymin, B.; Tenaud, C.; Coll, J.L. Intercellular trafficking and enhanced in vivo antitumour activity of a non-virally delivered P27-VP22 fusion protein. Gene Ther. 2003, 10, 314–325. [Google Scholar] [CrossRef] [PubMed]
  166. Zitvogel, L.; Pitt, J.M.; Daillere, R.; Smyth, M.J.; Kroemer, G. Mouse models in oncoimmunology. Nat. Rev. Cancer 2016, 16, 759–773. [Google Scholar] [CrossRef]
  167. Dexter, D.L.; Kowalski, H.M.; Blazar, B.A.; Fligiel, Z.; Vogel, R.; Gloria, H.; Heppner, H. Heterogeneity of Tumor Cells from a Single Mouse Mammary Tumor. Cancer Res. 1978, 38, 3174–3181. [Google Scholar]
  168. Heppner, G.H.; Dexter, D.L.; DeNucci, T.; Miller, F.R.; Calabresi, P. Heterogeneity in drug sensitivity among tumor cell subpopulations of a single mammary tumor. Cancer Res. 1978, 38, 3758–3763. [Google Scholar]
  169. Aslakson, C.J.; Miller, F.R. Selective Events in the Metastatic Process Defined by Analysis of the Sequential Dissemination of Subpopulations of a Mouse Mammary Tumor. Cancer Res. 1992, 52, 1399–1405. [Google Scholar]
  170. Yoneda, T.; Michigami, T.; Yi, B.; Williams, P.J.; Niewolna, M.; Hiraga, T. Actions of bisphosphonate on bone metastasis in animal models of breast carcinoma. Cancer 2000, 88, 2979–2988. [Google Scholar] [CrossRef]
  171. Guy, C.T.; Webster, M.A.; Schaller, M.; Parsons, T.J.; Cardiff, R.D.; Muller, W.J. Expression of the neu protooncogene in the mammary epithelium of transgenic mice induces metastatic disease. Proc. Natl. Acad. Sci. USA 1992, 89, 10578–10582. [Google Scholar] [CrossRef] [Green Version]
  172. Lollini, P.L.; De Giovanni, C.; Nanni, P. Preclinical HER-2 vaccines: From rodent to human HER-2. Front. Oncol. 2013, 3, 151. [Google Scholar] [CrossRef] [Green Version]
  173. Lollini, P.L.; Cavallo, F.; Nanni, P.; Forni, G. Vaccines for tumour prevention. Nat. Rev. Cancer 2006, 6, 204–216. [Google Scholar] [CrossRef]
  174. Sørlie, T.; Perou, C.M.; Tibshirani, R.; Aas, T.; Geisler, S.; Johnsen, H.; Hastie, T.; Eisen, M.B.; Van De Rijn, M.; Jeffrey, S.S.; et al. Gene expression patterns of breast carcinomas distinguish tumor subclasses with clinical implications. Proc. Natl. Acad. Sci. USA 2001, 98, 10869–10874. [Google Scholar] [CrossRef] [Green Version]
  175. Russnes, H.G.; Lingjærde, O.C.; Børresen-Dale, A.L.; Caldas, C. Breast Cancer Molecular Stratification: From Intrinsic Subtypes to Integrative Clusters. Am. J. Pathol. 2017, 187, 2152–2162. [Google Scholar] [CrossRef]
  176. Dobrolecki, L.E.; Airhart, S.D.; Alferez, D.G.; Aparicio, S.; Behbod, F.; Bentires-Alj, M.; Brisken, C.; Bult, C.J.; Cai, S.; Clarke, R.B.; et al. Patient-derived xenograft (PDX) models in basic and translational breast cancer research. Cancer Metastasis Rev. 2016, 35, 547–573. [Google Scholar] [CrossRef] [Green Version]
  177. Baker, K. Organoids provide an important window on inflammation in cancer. Cancers 2018, 10, 151. [Google Scholar] [CrossRef] [Green Version]
  178. Sachs, N.; de Ligt, J.; Kopper, O.; Gogola, E.; Bounova, G.; Weeber, F.; Balgobind, A.V.; Wind, K.; Gracanin, A.; Begthel, H.; et al. A Living Biobank of Breast Cancer Organoids Captures Disease Heterogeneity. Cell 2018, 172, 373–386. [Google Scholar] [CrossRef] [Green Version]
  179. Tuveson, D.; Clevers, H. Cancer modeling meets human organoid technology. Science 2019, 364, 952–955. [Google Scholar] [CrossRef]
  180. Du Manoir, S.; Orsetti, B.; Bras-Gonçalves, R.; Nguyen, T.T.; Lasorsa, L.; Boissière, F.; Massemin, B.; Colombo, P.E.; Bibeau, F.; Jacot, W.; et al. Breast tumor PDXs are genetically plastic and correspond to a subset of aggressive cancers prone to relapse. Mol. Oncol. 2014, 8, 431–443. [Google Scholar] [CrossRef]
  181. Dijkstra, K.K.; Cattaneo, C.M.; Weeber, F.; Chalabi, M.; van de Haar, J.; Fanchi, L.F.; Slagter, M.; van der Velden, D.L.; Kaing, S.; Kelderman, S.; et al. Generation of Tumor-Reactive T Cells by Co-culture of Peripheral Blood Lymphocytes and Tumor Organoids. Cell 2018, 174, 1586–1598. [Google Scholar] [CrossRef] [Green Version]
  182. Neal, J.T.; Li, X.; Zhu, J.; Giangarra, V.; Grzeskowiak, C.L.; Ju, J.; Liu, I.H.; Chiou, S.H.; Salahudeen, A.A.; Smith, A.R.; et al. Organoid Modeling of the Tumor Immune Microenvironment. Cell 2018, 175, 1972–1988. [Google Scholar] [CrossRef] [Green Version]
  183. Hu, J.; Sun, C.; Bernatchez, C.; Xia, X.; Hwu, P.; Dotti, G.; Li, S. T-cell homing therapy for reducing regulatory T cells and preserving effector T-cell function in large solid tumors. Clin. Cancer Res. 2018, 24, 2920–2934. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Origin of the TS/A cell line and variants. For pictures see [51].
Figure 1. Origin of the TS/A cell line and variants. For pictures see [51].
Cancers 11 01889 g001
Figure 2. Morphology of the TS/A cell line in adherent culture (phase contrast, ×100).
Figure 2. Morphology of the TS/A cell line in adherent culture (phase contrast, ×100).
Cancers 11 01889 g002
Table 1. TS/A model: main features.
Table 1. TS/A model: main features.
TopicsCell VariantsFeaturesRefs
Cytoskeletal markersE1CK8-positive[13]
Cytokine production TS/A, clones and variantsCSF[14,15,16,17]
TS/ATGF-β1 production (about 4 ng/mL)[18]
Cytokine receptorsTS/AIFN-γ receptor (1000/cell)[19]
Gene alterationsTS/AKaryotype[20]
TS/A and E1p53 mutated (codon 270 Arg to His)[13,21]
Gene expressionTS/ATERT (11,000 RNA copies)[22]
Hormone sensitivityTS/A and E1Estrogen receptor positive[10,13]
ImmunityTS/A Low immunogenicity[10,23,24]
TS/A and engineered variantsTumor associated antigen gp70env[25]
TS/A Suppressor activity[26,27]
TS/AMyeloid-derived suppressor cells (MDSC)[28,29,30,31,32,33,34,35]
TS/ANK resistance[36]
TS/AmD52 antigen[37]
Membrane moleculesTS/ACore 1 O-glycans[38]
TS/AMuc-1[39]
TS/ATag72[39]
PhenotypeTS/A, clones and variantsHeterogeneity (morphology, metastasis)[10,40,41]
Stem cell markersTS/ASca-1 (Ly6A/E)[42,43]
Tyrosine Kinase membrane receptorsTS/Ap185erbB2[39,44]
OthersTS/AEndoglin-negative[45]
TS/AFragile X mental retardation protein (FMRP), low expression[46]
TS/AHigh Mobility Group Box1 (HMGB1)-positive[47]
TS/ALats2[48]
TS/AST6Gal activity (present, low)[49]
TS/ATLR9-negative[50]
Table 2. TS/A in gene therapy studies aiming to increase tumor immunogenicity.
Table 2. TS/A in gene therapy studies aiming to increase tumor immunogenicity.
Immune CategoriesTransgenesVector/Transfer MethodsRefs
Chemokinesh-CCL16/LECNaked DNA + lipofection[70,71]
Adenoviral, in vivo[59,72]
Cytokinesm-IL2Naked DNA + electroporation[11,23,73,74,75,76,77]
m-IL4Retroviral[73,74,78,79,80,81,82,83,84,85]
m-IL5Retroviral[86,87]
m-IL6Retroviral[74,80,87]
m-IL7Naked DNA + electroporation[73,74,80,81,88,89]
m-IL7Adenoviral[90]
m-IL10Naked DNA + electroporation[74,91]
m-IL12 p35 and p40Naked DNA, in vivo[92,93]
Naked DNA + gene gun[94]
Naked DNA + lipofection[95]
Retroviral [96,97]
Canarypox[98]
Naked DNA + gene gun, in vivo[24]
h-IL13Naked DNA + calcium phosphate[99]
m-IL15Adenoviral[100]
IL15Naked DNA + lipofection[95,101]
Pro-IL18 and ICENaked DNA + gene gun[94]
m-IL21Naked DNA + lipofection[102]
m-IFNα1Naked DNA + electroporation[80,82,103,104,105,106]
m-IFNα1Naked DNA + gene gun[107]
m-IFNα4Naked DNA + polymer[108]
m-IFNβNaked DNA + calcium phosphate[109]
m-IFNγNaked DNA + lipofection[12,74,80,99,110,111,112,113]
m-GMCSFRetroviral[74]
m-TNFαRetroviral[74,80]
Membrane moleculesB7-1/CD80Naked DNA[114]
Naked DNA + electroporation[115,116]
Retroviral[81,89]
B7-2/CD86Naked DNA + electroporation[115,116]
Allogeneic MHCNaked DNA + calcium phosphate[111,117]
CD70 (CD27L)Retroviral[118,119]
CD153 (CD30L)Retroviral[118]
CD154 (CD40L)Retroviral[118,120]
TRAIL/APO2LNaked DNA + lipofection[121]
LAG-3 and LAG5Naked DNA + electroporation[122,123]
CCR7Naked DNA + calcium phosphate[124]
Suicide genesCytosine deaminaseRetroviral[112,125]
Naked DNA[126]
VSV (oncolytic)[127,128]
HSV-Thymidine kinaseNaked DNA[104]
Retroviral[129]
Naked DNA + lipofection[130]
OthersCIITANaked DNA + lipofection[131,132,133]
GBP1Retroviral (conditional) + naked DNA[134]
m-IRF1Adenoviral[135]
Table 3. TS/A in transduction studies of cancer biology.
Table 3. TS/A in transduction studies of cancer biology.
Gene CategoriesTransgenesVector/Transfer MethodsRefs
Oncosuppressorsm-p53wtCanarypox[21]
m-p53wt/mutVSV (oncolytic)[139]
Reporter genesLuciferaseNaked DNA[140,141]
β-galactosidaseNaked DNA + polyfection[142]
GFPAdenoviral[143]
GFPLentiviral[46]
EGFPLentiviral[54]
EGFPNaked DNA + electroporation[50]
EGFP (driven by p21 or CMV promoter)Naked DNA + electroporation[144,145]
Silencing antisense m-TGF-β1Retroviral[18]
Rab27aLentiviral[146]
Mlh1CRISPR-Cas9[147]
FoxP3Lentiviral siRNA[44]
fragile X mental retardation protein (FMRP)Lentiviral shRNA[46]
Surrogate antigensβ-galactosidaseRetroviral[81,148,149,150]
HemagglutininNaked DNA + lipofection[151,152]
Leishmania receptor for activated C kinase (LACK)Naked DNA[153,154,155]
Mycobacterial cell wall-associated 19-kDa lipoprotein [156]
OvalbuminNaked DNA [157,158]
OthersChromogranin A (Vasostatin-1 fragment)Naked DNA + electroporation[159,160,161]
Extracellular domain of receptor tyrosine kinase Tie2/TEK (ex-TEK)Naked DNA + calcium phosphate precipitation[162]
ApelinNaked DNA + polyfection[163]
Interferon-regulatory factor-1 (IRF-1)Adenoviral[43]
α1,2fucosyltransferaseNaked DNA + lipofection[164]
P27VP22Naked DNA + polyfection[165]

Share and Cite

MDPI and ACS Style

De Giovanni, C.; Nicoletti, G.; Landuzzi, L.; Palladini, A.; Lollini, P.-L.; Nanni, P. Bioprofiling TS/A Murine Mammary Cancer for a Functional Precision Experimental Model. Cancers 2019, 11, 1889. https://doi.org/10.3390/cancers11121889

AMA Style

De Giovanni C, Nicoletti G, Landuzzi L, Palladini A, Lollini P-L, Nanni P. Bioprofiling TS/A Murine Mammary Cancer for a Functional Precision Experimental Model. Cancers. 2019; 11(12):1889. https://doi.org/10.3390/cancers11121889

Chicago/Turabian Style

De Giovanni, Carla, Giordano Nicoletti, Lorena Landuzzi, Arianna Palladini, Pier-Luigi Lollini, and Patrizia Nanni. 2019. "Bioprofiling TS/A Murine Mammary Cancer for a Functional Precision Experimental Model" Cancers 11, no. 12: 1889. https://doi.org/10.3390/cancers11121889

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop