Skip to main content
Cancer Science logoLink to Cancer Science
. 2022 Apr 18;113(6):1919–1929. doi: 10.1111/cas.15369

Tumor‐targeted fluorescence labeling systems for cancer diagnosis and treatment

Hiroshi Tazawa 1,2,, Kunitoshi Shigeyasu 1, Kazuhiro Noma 1, Shunsuke Kagawa 1,3, Fuminori Sakurai 4, Hiroyuki Mizuguchi 4, Hisataka Kobayashi 5, Takeshi Imamura 6, Toshiyoshi Fujiwara 1
PMCID: PMC9207361  PMID: 35398956

Abstract

Conventional imaging techniques are available for clinical identification of tumor sites. However, detecting metastatic tumor cells that are spreading from primary tumor sites using conventional imaging techniques remains difficult. In contrast, fluorescence‐based labeling systems are useful tools for detecting tumor cells at the single‐cell level in cancer research. The ability to detect fluorescent‐labeled tumor cells enables investigations of the biodistribution of tumor cells for the diagnosis and treatment of cancer. For example, the presence of fluorescent tumor cells in the peripheral blood of cancer patients is a predictive biomarker for early diagnosis of distant metastasis. The elimination of fluorescent tumor cells without damaging normal tissues is ideal for minimally invasive treatment of cancer. To capture fluorescent tumor cells within normal tissues, however, tumor‐specific activated target molecules are needed. This review focuses on recent advances in tumor‐targeted fluorescence labeling systems, in which indirect reporter labeling using tumor‐specific promoters is applied to fluorescence labeling of tumor cells for the diagnosis and treatment of cancer. Telomerase promoter‐dependent fluorescence labeling using replication‐competent viral vectors produces fluorescent proteins that can be used to detect and eliminate telomerase‐positive tumor cells. Tissue‐specific promoter‐dependent fluorescence labeling enables identification of specific tumor cells. Vimentin promoter‐dependent fluorescence labeling is a useful tool for identifying tumor cells that undergo epithelial–mesenchymal transition (EMT). The evaluation of tumor cells undergoing EMT is important for accurately assessing metastatic potential. Thus, tumor‐targeted fluorescence labeling systems represent novel platforms that enable the capture of tumor cells for the diagnosis and treatment of cancer.

Keywords: adenovirus, EMT, survivin, telomerase, vimentin


This review focuses on recent advances in tumor‐targeted fluorescence labeling systems. Indirect reporter labeling induces the expression of genes encoding fluorescent proteins in tumor cells using tumor‐specific gene promoters.

graphic file with name CAS-113-1919-g003.jpg


Abbreviations

Ad5

adenovirus type 5

Ad35

adenovirus type 35

AFP

alpha‐fetoprotein

CAR

coxsackievirus and adenovirus receptor

CEA

carcinoembryonic antigen

CTC

circulating tumor cell

ctDNA

cell‐free tumor DNA

EGFP

enhanced green fluorescent protein

EMT

epithelial–mesenchymal transition

EV

extracellular vesicle

FSP1

fibroblast‐specific protein 1

GFP

green fluorescent protein

HCC

hepatocellular carcinoma

HSV1

human simplex virus type‐1

hTERT

human telomerase reverse transcriptase

Id1

inhibitor of DNA binding 1

IL‐1β

interleukin‐1β

miRNA

microRNA

PDAC

pancreatic ductal adenocarcinoma

PDT

photodynamic therapy

PSA

prostate‐specific antigen

PSES

prostate‐specific enhancer sequence

PSMA

prostate‐specific membrane antigen

RFP

red fluorescent protein

ROS

reactive oxygen species

SEAP

secreted embryonic alkaline phosphatase

TME

tumor microenvironment

1. INTRODUCTION

Tumor cells exhibit characteristics that increase the potential for malignancy, such as unlimited cell proliferation, chemoresistance, migratory and invasive abilities, and metastasis, which can lead to poor prognosis in cancer patients. Recent advances in antitumor techniques such as surgery, chemotherapy, radiotherapy, and immunotherapy have improved the clinical outcome of cancer patients. However, advanced cancers are often refractory to antitumor therapies and exhibit tumor recurrence, distant metastasis, and poor prognosis. Therefore, early diagnosis of cancer is one of the most prominent strategies to cure cancer patients.

A variety of medical imaging techniques are available to detect tumor sites in clinical practice, including X‐rays, ultrasound, computed tomography, and magnetic resonance imaging. These techniques enable the detection of primary and metastatic tumors throughout the body. 1 Nuclear medicine techniques, including PET and single photon emission computed tomography, can be used in supportive fashion to confirm the malignant potential of tumor tissue using biomolecule‐targeted radioactive isotopes. 1 These imaging techniques are useful for determining the anatomic localization of tumors and their invasiveness into surrounding normal tissues. However, detecting invasive and metastatic tumor cells that are spreading from primary tumors remains difficult using conventional imaging techniques. Therefore, novel imaging techniques useful for evaluating the biodistribution of metastatic tumor cells are needed.

Fluorescence‐based labeling systems are powerful tools for detecting tumor cells at the single‐cell level in cancer research. Various fluorescent proteins, including GFP and RFP, are widely used to visualize the behavior of tumor cells using intravital imaging techniques in animal experiments. 2 The detection of tumor cells using these fluorescence labeling systems enables investigations of the biodistribution of tumor cells for the diagnosis and treatment of cancer. For example, the presence of CTCs in the peripheral blood of cancer patients is a predictive biomarker for early diagnosis of distant metastasis. Liquid biopsy has recently emerged as a minimally invasive procedure for the detection of CTCs, 3 ctDNA, 3 miRNA, 4 and EVs 5 in body fluids. In contrast, the ability to eliminate tumor cells without damaging normal tissues would be ideal in minimally invasive treatment of cancer. Oncolytic virotherapy using tumor‐specific replication‐competent viruses has recently emerged as a novel antitumor technique to induce tumor‐specific cell death without harming normal cells. 6 Replication‐competent viral vectors carrying tumor‐specific gene promoters can be further applied to the fluorescence labeling of CTCs in the peripheral blood of cancer patients. 7 Thus, fluorescence labeling systems are useful options for identifying tumor cells in the diagnosis and treatment of cancer.

2. TUMOR‐TARGETED FLUORESCENCE LABELING SYSTEMS

Indirect reporter labeling is one of the leading tumor‐targeted fluorescence labeling systems that contribute to the diagnosis and treatment of tumors (Figure 1). Indirect reporter labeling induces the expression of genes encoding fluorescent proteins in tumor cells using tumor‐specific gene promoters, which is useful for the diagnosis of cancer. Tumor‐specific activated molecules are used to identify tumor cells in tumor‐targeted fluorescence labeling systems. There are several types of intrinsic factors that are activated in a tumor‐specific manner and related to malignancy potential. Unlimited cell proliferation is the most common feature of the vast majority of tumor cells, in which telomerase activity plays a central role in preventing replicative senescence through elongation of telomeres. 8 Activation of antiapoptotic survival pathways, such as survivin 9 and Id1, 10 inevitably contributes to chemoresistance. Detecting tumor markers such as PSA, CEA, and AFP in the serum of cancer patients is useful in the assessment of tissue‐specific tumors. 11 Migration and invasiveness are well‐known characteristics of metastatic tumor cells in association with EMT, which is a fundamental process that induces the mesenchymal phenotype in epithelial tumor cells. 12 Epithelial–mesenchymal signatures in tumor tissues are highly associated with poor prognosis in patients with various types of cancers. 13 , 14 , 15 , 16 Given the tumor‐promoting role of various intrinsic factors, these factors could be exploited as tumor‐specific target molecules to identify tumor cells in the diagnosis and treatment of cancer.

FIGURE 1.

FIGURE 1

Tumor‐targeted fluorescence labeling systems using indirect reporters. EMT, epithelial–mesenchymal transition

Optical imaging techniques are needed to detect fluorescently labeled tumor cells in vivo. As several other reviews have summarized in vivo optical imaging techniques, 17 , 18 we focus here on fluorescence‐based labeling systems that enable identification of tumor cells through the targeting of tumor‐specific molecules. We summarize various types of fluorescence labeling systems based on tumor‐specific activated molecules, such as telomerases, survivin, tumor markers, and the EMT process. Moreover, we discuss future perspectives regarding the clinical application of tumor‐targeted fluorescence labeling systems for the diagnosis and treatment of cancer.

3. TUMOR‐SPECIFIC PROMOTER‐DEPENDENT FLUORESCENCE LABELING

Indirect reporter labeling induces the expression of fluorescent proteins in tumor cells using tumor‐specific promoters (Figure 1). There are several types of tumor‐specific gene promoters that are used to regulate the expression of therapeutic transgenes in cancer gene therapy. 19 These tumor‐specific gene promoters can be applied to the fluorescent labeling of tumor cells. Replication‐competent viral vectors carrying tumor‐specific gene promoters are useful tools for inducing the expression of fluorescent proteins in tumor cells. 20

3.1. Telomerase promoter‐dependent fluorescence labeling

Telomerase is an enzyme that elongates the length of telomeres by repeated TTAGGG nucleotide sequences at the ends of a chromosome. 21 Most tumor cells exhibit high telomerase activity in association with unlimited cell proliferation, whereas most types of normal somatic cells exhibit no telomerase activity. Telomerase is a ribonucleoprotein complex consisting of two different subunits, a catalytic subunit (hTERT and telomerase‐associated protein 1) 22 , 23 and an RNA subunit (human telomerase RNA component). 24 As hTERT expression is highly associated with telomerase activity in tumor cells, 25 , 26 we developed a telomerase‐dependent replicative adenovirus, OBP‐401 (TelomeScan), in which the hTERT gene promoter is inserted into the Ad5 genome to drive the expression of the adenoviral E1A and E1B genes for tumor cell‐specific viral replication 27 (Figure 2 and Table 1). OBP‐401 harbors the GFP expression cassette in the E3 region, enabling visualization of tumor cells as GFP‐positive cells (Figure 2). The OBP‐401‐based GFP reporter labeling system is useful for the detection of CTCs in the peripheral blood. 28 When GFP‐positive CTCs isolated by flow cytometry were analyzed using direct sequencing and allele‐specific PCR, the mutation status in the KRAS/BRAF oncogenes was similarly detectable between primary tumor tissues and CTCs in patients with colorectal cancers. 29 Moreover, when intraoperative peritoneal wash collected for conventional cytology was infected with OBP‐401, OBP‐401 induced GFP‐positive peritoneal tumor cells, which are associated with poor prognosis, especially in cytology‐positive patients with gastric cancers. 30

FIGURE 2.

FIGURE 2

Fluorescence labeling systems using telomerase promoter‐dependent replicative viruses expressing fluorescent proteins. Ad5, adenovirus type 5; Ad35, adenovirus type 35; CMV, cytomegalovirus; GFP, green fluorescent protein; hTERT, human telomerase reverse transcriptase; IRES, internal ribosome entry site; ITR, inverted terminal repeat; miR, microRNA

TABLE 1.

Summary of tumor‐specific or tissue‐specific promoter‐dependent adenoviruses expressing fluorescent proteins

No. Promoter Virus Origin Fluorescent protein Fiber Application References
1 hTERT OBP‐401 (TelomeScan) Ad5 GFP Ad5 Detection of CTC and DTC 25, 26, 27, 28
2 hTERT OBP‐1101 (TelomeScan F35) Ad5 GFP Ad35 Detection of CTC 30, 34
3 hTERT TelomeKiller Ad5 KillerRed Ad5 Elimination of tumor cell 44, 45
4 Survivin Ad5/35E1apsurvivinE4 Ad5 GFP Ad35 Detection of CTC 47
5 Survivin AdSur‐SYE Ad5 EGFP SYENFSA ligand Detection of PDAC cell 48, 49
6 Survivin CRAd5/11‐Sp‐eGFP Ad5 EGFP Ad5/11 Elimination of glioma cell 50
7 Id1 Ad5/3‐Id1‐SEAP‐Id1‐mCherry Ad5 mCherry Ad5/3 Detection of tumor cell 53, 54
8 PSES Ad5/35E1aPSESE4 Ad5 GFP Ad35 Detection of PC cell 55, 56
9 CEA AdCEAp‐EGFP Ad5 EGFP Ad5 Detection of PDAC cell 59
10 AFP Ad/Ha2bm‐GFP Ad5 GFP Ad5 Detection of HCC cell 63

Abbreviations: Ad5, adenovirus type 5; Ad35, adenovirus type 35; AFP, alpha‐fetoprotein; CEA, carcinoembryonic antigen; CTC, circulating tumor cell; DTC, disseminated tumor cell; EGFP, enhanced green fluorescent protein; GFP, green fluorescent protein; HCC, hepatocellular carcinoma; hTERT, human telomerase reverse transcriptase; Id1, inhibitor of DNA binding 1; PC, prostate cancer; PDAC, pancreatic ductal adenocarcinoma; PSES, prostate‐specific enhancer sequence.

Although the OBP‐401‐based fluorescence labeling system is useful for detecting telomerase‐positive tumor cells, there are some limitations to this labeling system. One limitation is the possibility of false‐positive detection of populations of hematopoietic cells, such as CD13+ CD14+ monocytes, which are detected as GFP‐positive cells in the peripheral blood. 31 Another limitation is false‐negative results due to missed detection of populations of tumor cells lacking CAR expression. To address these limitations, Sakurai et al. 32 developed a modified OBP‐401 (OBP‐1101, TelomeScan F35), in which the target sequences of miR‐142‐3p are inserted into the E1 and E3 regions and the fiber of Ad5 is replaced with that of Ad35 (Figure 2 and Table 1). As miR‐142‐3p is ubiquitously expressed in various types of hematopoietic cells, including monocytes, 33 , 34 OBP‐1101‐mediated E1 and GFP expression is attenuated in miR‐142‐3p‐positive hematopoietic cells, 32 leading to an improvement in tumor‐specific fluorescent labeling. Although Ad5 can bind to CAR on the surface of target cells, 35 Ad35 binds with greater affinity to CD46, which is expressed on a variety of tumor cells more ubiquitously than CAR. OBP‐1101 with Ad35 fibers can infect CAR‐negative tumor cells by binding to CD46, 32 leading to an improvement in viral infectivity. Togo et al. 36 reported that the OBP‐1101‐based fluorescence labeling system is a promising tool for detecting highly malignant CTCs expressing the mesenchymal marker vimentin in non‐small‐cell lung cancer patients. OBP‐1101 thus appears to be superior to OBP‐401 in terms of inducing tumor‐specific GFP expression.

Herpes simplex virus type‐1 can be also utilized as a tool to induce tumor‐specific GFP expression using the hTERT promoter. Zhang et al. 37 developed a telomerase‐dependent replicative herpes simplex virus, oHSV1‐hTERT‐GFP, in which the endogenous ICP4 promoter is replaced with the hTERT gene promoter to drive the expression of ICP4 for tumor‐specific viral replication (Figure 2 and Table 1). oHSV1‐hTERT‐GFP contains the GFP expression cassette in the ICP34.5 region, thus enabling visualization of tumor cells as GFP‐positive cells. The oHSV‐hTERT‐GFP‐based GFP induction system is useful for detecting CTCs in the peripheral blood of cancer patients. 38

Human telomerase reverse transcriptase promoter‐driven replicative viruses have emerged as not only novel diagnostic tools but also novel antitumor methods for cancer treatment. 39 OBP‐301 (suratadenoturev), the original virus of OBP‐401, exhibits a broad spectrum of antitumor efficacy against malignant tumor cells with telomerase activity. 40 OBP‐401 also exhibits antitumor effects against a variety of cancers, including lung cancer, 41 colon cancer, 41 breast cancer, 42 and gastric cancer. 43 Intraperitoneal injection of OBP‐401 was shown to enhance the therapeutic efficacy of chemotherapeutic agents in mice with intraperitoneal metastasis of gastric cancer. 43 In contrast, PDT is a minimally invasive antitumor technique for inducing tumor‐specific cytotoxicity through the induction of photosensitizer‐mediated generation of ROS upon light irradiation. 44 , 45 For the application of hTERT promoter‐driven replicative viruses to PDT, we generated a telomerase‐specific replicative photodynamic viral agent known as TelomeKiller, which harbors the KillerRed expression cassette in the E3 region, enabling elimination of tumor cells through KillerRed‐mediated generation of ROS upon green‐light irradiation 46 , 47 (Figure 2 and Table 1). Thus, hTERT promoter‐driven viral vectors are useful tools for detecting and eradicating telomerase‐positive tumor cells.

3.2. Survivin promoter‐dependent fluorescence labeling

Survivin is an inhibitor of apoptosis and plays a central role in the survival of cancer cells by inhibiting therapy‐induced cell death. 9 Survivin expression is highly upregulated in a variety of tumor cells at the transcriptional level, 9 suggesting that the survivin gene promoter is a universal tumor‐specific promoter, similar to the hTERT gene promoter. Kamizono et al. 48 showed that the survivin promoter is useful for the tumor‐specific replication of oncolytic adenoviruses. For the fluorescent labeling of survivin‐positive tumor cells, Seo et al. 49 developed a survivin promoter‐specific replicative adenovirus, Ad5/35E1apsurvivinE4, in which the survivin promoter is inserted into the Ad5 genome to drive expression of the adenoviral E1A and E4 genes for survivin‐specific virus replication (Figure 3 and Table 1). Ad5/35E1apsurvivinE4 encodes the GFP expression cassette and Ad35 fiber gene, thus enabling visualization of survivin‐positive cancer cells independent of CAR expression (Figure 3). In contrast, Yamamoto et al. 50 generated a survivin promoter‐driven replicative adenovirus, AdSur‐SYE, in which the E1 promoter is replaced with survivin promoter for survivin‐specific virus replication (Figure 3 and Table 1). The fiber of AdSur‐SYE contains the SYENFSA ligand, which is sequence identified to bind with greater affinity to human pancreatic cancer cells. 51 As AdSur‐SYE harbors the EGFP expression cassette in the E3 region, the AdSur‐SYE‐based EGFP induction system is highly effective for targeting pancreatic cancer cells. In addition, Li et al. 52 developed a survivin promoter‐specific replicative adenovirus, CRAd5/11‐Sp‐eGFP, which expresses EGFP and chimeric Ad5/11 fiber consisting of an Ad5 tail and an Ad11 shaft and knob (Figure 3 and Table 1). CRAd5/11‐Sp‐eGFP efficiently detects and eliminates survivin‐positive glioma cells. Survivin promoter‐driven viral vectors are thus useful tools for the detection and elimination of survivin‐positive tumor cells.

FIGURE 3.

FIGURE 3

Fluorescence labeling systems using survivin promoter‐dependent replicative viruses expressing fluorescent proteins. Ad5, adenovirus type 5; Ad35, adenovirus type 35; CMV, cytomegalovirus; EGFP, enhanced green fluorescent protein; GFP, green fluorescent protein; hTERT, human telomerase reverse transcriptase; IRES, internal ribosome entry site; ITR, inverted terminal repeat; miR, microRNA

3.3. Inhibitor of DNA binding 1 promoter‐dependent fluorescence labeling

Inhibitor of DNA binding 1 is a member of the helix–loop–helix transcription factor family of proteins that control a variety of cellular processes, including cell proliferation and cell cycle regulation. 53 As expression of Id1 mRNA and protein is increased in multiple types of cancer, 54 the Id1 promoter is a promising candidate for a universal tumor‐specific promoter. Warram et al. 55 developed a dual reporter adenoviral vector, Ad5/3‐Id1‐SEAP‐Id1‐mCherry, in which the Id1 promoter is inserted into the E1‐ and E3‐deleted Ad5 genome to drive expression of the SEAP enzyme for blood‐based screening and the fluorescent reporter mCherry for detecting Id1‐positive tumor cells (Table 1). Ad5/3‐Id1‐SEAP‐Id1‐mCherry expresses a hybrid Ad5/3 fiber for improved infectivity of CAR‐negative tumor cells by binding to CD46. Infection with Ad5/3‐Id1‐SEAP‐Id1‐mCherry results in the secretion of SEAP and mCherry by human breast and prostate cancer cells. 55 , 56 This dual reporter system is a unique and useful approach for detecting Id1‐positive tumor cells using blood‐based screening and tumor visualization.

4. TISSUE‐SPECIFIC PROMOTER‐DEPENDENT FLUORESCENCE LABELING

Tissue‐specific promoters are useful for inducing the expression of fluorescent proteins in some tumor cells (Figure 1). Tissue‐specific gene promoters are used to drive the expression of therapeutic transgenes in cancer gene therapy. 19 Replication‐competent viral vectors with tissue‐specific gene promoters are also useful tools for inducing the expression of fluorescent proteins in tumor cells. 20

4.1. Prostate‐specific promoter‐dependent fluorescence labeling

Prostate‐specific antigen and PSMA are highly sensitive markers for prostate cancer. To target prostate cancer cells in gene therapy, Lee et al. 57 developed a PSES, which is a chimeric enhancer derived from the promoter of the PSA and PSMA genes. For the fluorescent labeling of prostate cancer cells, Hwang et al. 58 developed a prostate‐specific replicative adenovirus, Ad5/35E1aPSESE4, in which the PSES gene is inserted into the Ad5 genome to drive expression of the adenoviral E1A and E4 genes for prostate‐specific virus replication (Table 1). Ad5/35E1aPSESE4 encodes the GFP expression cassette and Ad35 fiber gene, thereby enabling visualization of prostate cancer cells independent of CAR expression (Table 1). The Ad5/35E1aPSESE4‐based GFP induction system is useful for detecting CTCs in the peripheral blood of prostate cancer patients. 58 Prostate promoter‐driven viral vectors are thus useful tools for detecting prostate tumor cells.

4.2. Carcinoembryonic antigen promoter‐dependent fluorescence labeling

Pancreatic ductal adenocarcinoma is a highly lethal disease, with a 5‐year survival rate of less than 10%. 59 Tools enabling early diagnosis of PDAC are needed to improve the clinical outcome of PDAC patients. Carcinoembryonic antigen is the standard serum tumor marker for assessing a variety of gastrointestinal cancers, including pancreatic cancer. 60 The secretion of CEA to the extracellular environment is highly associated with CEA promoter activity in PDAC cells. 61

To target CEA‐positive PDAC cells, Xu et al. 61 developed CEA promoter‐specific replicative adenoviruses expressing EGFP (AdCEAp‐EGFP) or heat shock protein 70 (AdCEAp‐Hsp70), in which the CEA promoter‐driven E1A expression cassettes are inserted into the Ad5 genome (Table 1). AdCEAp‐EGFP induces EGFP expression to enable visualization of CEA‐positive PDAC cells, whereas AdCEAp‐Hsp70 induces cytopathic effects useful in the treatment of CAE‐positive PDAC cells. Thus, CEA promoter‐driven viral vectors are promising tools for detecting and eliminating CEA‐positive PDAC cells.

4.3. Alpha‐fetoprotein promoter‐dependent fluorescence labeling

Hepatocellular carcinoma is the most common type of liver cancer. 62 Alpha‐fetoprotein is widely used as serum tumor marker for the diagnosis of HCC. 63 Alpha‐fetoprotein expression is highly associated with clinical stage, early recurrence, and poor prognosis in HCC patients. 64 For the fluorescence labeling of AFP‐positive HCC cells, Yoon et al. 65 developed an AFP promoter‐specific nonreplicative adenovirus carrying the GFP gene (Ad/Ha2bm‐GFP) (Table 1). Ad/Ha2bm‐GFP encodes the GFP expression cassette under the control of a modified AFP promoter containing two types of enhancer regions and hypoxia‐responsive elements. Ad/Ha2bm‐GFP efficiently induces GFP expression in AFP‐positive HCC cells even under hypoxic conditions. Thus, AFP promoter‐driven viral vectors are useful for detecting AFP‐positive HCC cells.

5. EPITHELIAL–MESENCHYMAL TRANSITION‐DEPENDENT FLUORESCENCE LABELING

Invasion and metastasis are hallmarks of cancer. The processes of invasion and metastasis are highly associated with the EMT program, which is a fundamental process by which epithelial tumor cells acquire mesenchymal characteristics with invasive and metastatic potential. 12 In tumor cells undergoing EMT, mesenchymal markers such as N‐cadherin and vimentin are upregulated, whereas epithelial markers such as E‐cadherin and cytokeratin are downregulated. The EMT program is cooperatively regulated by several intrinsic factors, including EMT‐activating transcription factors and EMT‐suppressive miRNAs. 66 As EMT signatures are highly associated with poor prognosis in patients with a variety of cancers, 13 , 14 , 15 , 16 the evaluation of tumor cells undergoing EMT is important to accurately assess metastatic potential. Indirect reporter labeling systems based on EMT‐dependent promoters are available to induce the expression of fluorescent proteins in tumor cells undergoing EMT (Figure 1).

5.1. Irreversible EMT‐dependent fluorescence labeling

For the fluorescence labeling of tumor cells undergoing EMT, Fischer et al. 67 developed a transgenic mouse model using a fibroblast‐specific marker‐dependent fluorescence switching system, in which the FSP1 promoter drives the Cre‐lox recombinase system to induce the RFP‐to‐GFP conversion in tumor cells undergoing EMT (Figure 4). Zhao et al. 68 used transgenic mice with the FSP1‐dependent fluorescence switching system to demonstrate that tumor cells undergoing EMT localize in the area of tumors close to blood vessels. In contrast, in experiments using the FSP1‐dependent fluorescence switching system, Bornes et al. reported that a subpopulation of pre‐EMT RFP‐positive cells with low epithelial marker expression exhibited greater metastatic potential than post‐EMT GFP‐positive cells. 69 Lourenco et al. 70 also showed that pre‐EMT RFP‐positive cells play a more dominant role in lung metastasis than post‐EMT GFP‐positive cells. Recent evidence also implicates partial/hybrid EMT cells that possess both epithelial and mesenchymal characteristics due to EMT plasticity in the metastatic process. 66 , 71 As Cre‐lox recombinase‐based fluorescence labeling is irreversible, this technique might be inadequate for the real‐time labeling of EMT plasticity in tumor cells.

FIGURE 4.

FIGURE 4

Fluorescence labeling systems using epithelial–mesenchymal transition‐specific promoter‐dependent fluorescent probes. EMT, epithelial‐mesenchymal transition; FSP1, fibroblast‐specific protein 1; GFP, green fluorescent protein; IL‐1β, interleukin‐β; miR, microRNA; RFP, red fluorescent protein; TNF‐α, tumor necrosis factor‐α; UTR, untranslated region; VIM, vimentin

5.2. Reversible EMT‐dependent fluorescence labeling

The plasticity of the EMT program makes it difficult to detect tumor cells undergoing EMT during the metastasis process. Therefore, fluorescence labeling systems that enable reversible visualization of tumor cells undergoing EMT are needed. To investigate the plasticity of the EMT program in tumor cells, we developed a novel fluorescence labeling system using the VRV3 vector, in which the vimentin promoter‐dependent RFP expression cassette encodes the 3′‐UTR of vimentin 72 (Figure 4). The vimentin promoter‐dependent RFP expression vector lacking the 3′‐UTR of vimentin induces RFP expression in non‐EMT tumor cells, whereas addition of the 3′‐UTR of vimentin enables reversible visualization of EMT‐dependent RFP expression in human colorectal cancer HCT116 and RKO cells. 72 Treatment with inflammatory cytokines, tumor necrosis factor‐α, and IL‐1β induced EMT‐related RFP expression in HCT116‐VRV3 and RKO‐VRV3 cells (Figure 4). Conversely, removal of inflammatory cytokines reduced RFP expression in HCT116‐VRV3 and RKO‐VRV3 cells, indicating that this labeling system is reversible. In vivo experiments using HCT116‐VRV3 cells indicated that RFP‐positive tumor cells undergoing EMT localized within primary and metastatic tumor areas harboring IL‐1β‐positive inflammatory macrophages. 72 Consistent with HCT116‐VRV3 and RKO‐VRV3 cells, the ATCC recently established several EMT‐reporter cell lines, including A549 VIM RFP (CCL‐185EMT), HCT116 VIM RFP (CCL‐247EMT), and MD‐MB‐231 VIM RFP (HTB‐26EMT). These cells were generated by inserting the RFP reporter gene before the stop codon in the last exon of the vimentin gene using genome editing 73 (Figure 4). Treatment with miR‐200 inhibitor induced expression of the VIM‐RFP fusion protein in HCT116 VIM RFP cells (Figure 4). These vimentin promoter‐dependent fluorescence labeling systems are useful tools for exploring the underlying mechanism of EMT regulation in tumor cells.

6. FUTURE PERSPECTIVES

Telomerase‐targeted fluorescence labeling systems using viral vectors are useful tools for detecting tumor cells in the peripheral blood of cancer patients. Although the presence of CTCs is a predictive biomarker for the early diagnosis of distant metastasis, 3 it can be difficult to detect the small population of tumor cells in all cancer patients. Several types of biomarkers, such as ctDNA, 3 miRNA, 4 and EVs, 5 have recently emerged as liquid biopsy markers to evaluate the malignant potential of cancer patients. Recent reports have suggested that the premetastatic niche, which is a metastasis‐supportive microenvironment, is established by primary tumor‐derived EVs in secondary organs before the metastatic colonization by CTCs. 74 Assessing both CTCs and EVs could be more useful for the surveillance of metastatic potential in all cancer patients.

Tumor‐targeted replicative viruses expressing fluorescent proteins enable the visualization of tumor cells within normal tissues, which enhances the eradication of tumor cells when combined with various antitumor therapies. Intratumoral injection of the telomerase‐specific replicative adenovirus OBP‐401 induces the fluorescence labeling of primary tumors and tumor cells metastasized to lymph nodes, which facilitates the surgical resection of tumor areas using fluorescence‐guided surgery, thereby preventing tumor recurrence. 75 , 76 In contrast, intraperitoneal administration of OBP‐401 enables the fluorescence labeling of intraperitoneally disseminated tumor cells, which enhances the sensitivity to chemotherapeutic agents by induction of mitotic catastrophe. 43 Therefore, tumor‐specific replicative viruses expressing fluorescent proteins could provide promising options to treat fluorescent‐labeled tumor cells by combining antitumor techniques.

Vimentin‐targeted fluorescence labeling systems offer opportunities to investigate the behavior of tumor cells undergoing EMT during tumor progression. The detection of tumor cells undergoing EMT is suggestive of the involvement of EMT‐inducing factors within surrounding normal tissues. Takahashi et al. 77 recently reported a tissue‐clearing technology that is useful for visualizing the cell cycle status of primary and metastatic tumor cells in nude mice using human cancer cells stably expressing a fluorescent ubiquitination‐based cell cycle indicator. This tissue‐clearing method could be useful for analyzing the biodistribution of metastatic tumor cells undergoing EMT in mice using vimentin‐targeted fluorescence labeling systems. In contrast, although EMT features are highly associated with poor prognosis in cancer patients, efforts to develop anti‐EMT therapies have thus far been unsuccessful. Zhao et al. 78 recently reported that 3D organoid culture systems using mesenchymal breast cancer are useful for the screening of EMT‐reversing drugs by evaluating the resulting morphological changes. Using vimentin promoter‐dependent fluorescence labeling systems, it is possible to identify EMT‐inducing factors within the TME and thereby candidate reagents for anti‐EMT therapy. Vimentin promoter‐dependent fluorescence labeling systems could be useful options for evaluating the potential of both EMT‐inducing factors and EMT‐inhibiting drugs.

7. CONCLUSIONS

Fluorescence‐based labeling systems using several types of tumor‐specific target molecules enable the visualization of tumor cells as a means of investigating their biodistribution within normal tissues. The detection of metastatic tumor cells in the peripheral blood of cancer patients is a predictive biomarker for the surveillance of early metastasis. The characterization of EMT tumor cells could facilitate elucidation of the underlying mechanism of metastatic potential acquisition in association with the TME. The development of novel antitumor techniques using tumor‐specific target molecules is an ideal minimally invasive strategy to treat cancer patients without affecting normal tissues. Although a number of obstacles remain in investigating the biodistribution of tumor cells in the human body using optical imaging techniques, tumor‐targeted fluorescence labeling systems are promising novel platforms for detecting tumor cells in the diagnosis and treatment of cancer.

CONFLICTS OF INTEREST

Dr. Imamura Takeshi is an editorial board member of Cancer Science. The other authors have no potential conflicts of interest to disclose.

ACKNOWLEDGMENTS

This study was supported in part by JSPS KAKENHI grants JP21K07219 to H. Tazawa, JP20K09009 and JP24791423 to K. Noma, JP23591932 to S. Kagawa, JP20H00664 to H. Mizuguchi, and JP22390256 and JP16H05416 to T. Fujiwara. H. Kobayashi was supported by the Intramural Research Program of the National Institutes of Health, National Cancer Institute (ZIA BC 011513).

Tazawa H, Shigeyasu K, Noma K, et al. Tumor‐targeted fluorescence labeling systems for cancer diagnosis and treatment. Cancer Sci. 2022;113:1919–1929. doi: 10.1111/cas.15369

Funding information

Japan Society for the Promotion of Science KAKENHI, Grant/Award Number: JP21K07219, JP20K09009, JP24791423, JP23591932, JP20H00664, JP22390256, JP16H05416; NIH, NCI, Grant/Award Number: ZIA BC 011513

REFERENCES

  • 1. Barentsz J, Takahashi S, Oyen W, et al. Commonly used imaging techniques for diagnosis and staging. J Clin Oncol. 2006;24:3234‐3244. [DOI] [PubMed] [Google Scholar]
  • 2. Hoffman RM. Use of fluorescent proteins and color‐coded imaging to visualize cancer cells with different genetic properties. Cancer Metastasis Rev. 2016;35:5‐19. [DOI] [PubMed] [Google Scholar]
  • 3. Alix‐Panabieres C, Pantel K. Clinical applications of circulating tumor cells and circulating tumor DNA as liquid biopsy. Cancer Discov. 2016;6:479‐491. [DOI] [PubMed] [Google Scholar]
  • 4. Shigeyasu K, Toden S, Zumwalt TJ, Okugawa Y, Goel A. Emerging role of MicroRNAs as liquid biopsy biomarkers in gastrointestinal cancers. Clin Cancer Res. 2017;23:2391‐2399. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5. Yoshioka Y, Katsuda T, Ochiya T. Extracellular vesicles and encapusulated miRNAs as emerging cancer biomarkers for novel liquid biopsy. Jpn J Clin Oncol. 2018;48:869‐876. [DOI] [PubMed] [Google Scholar]
  • 6. Zheng M, Huang J, Tong A, Yang H. Oncolytic viruses for cancer therapy: barriers and recent advances. Mol Ther Oncolytics. 2019;15:234‐247. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7. Cao GD, He XB, Sun Q, et al. The oncolytic virus in cancer diagnosis and treatment. Front Oncol. 2020;10:1786. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 8. Hahn WC, Stewart SA, Brooks MW, et al. Inhibition of telomerase limits the growth of human cancer cells. Nat Med. 1999;5:1164‐1170. [DOI] [PubMed] [Google Scholar]
  • 9. Yamamoto H, Ngan CY, Monden M. Cancer cells survive with survivin. Cancer Sci. 2008;99:1709‐1714. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10. Zhang X, Ling MT, Wong YC, Wang X. Evidence of a novel antiapoptotic factor: role of inhibitor of differentiation or DNA binding (Id‐1) in anticancer drug‐induced apoptosis. Cancer Sci. 2007;98:308‐314. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11. Amayo AA, Kuria JG. Clinical application of tumour markers: a review. East Afr Med J. 2009;86:S76‐83. [DOI] [PubMed] [Google Scholar]
  • 12. Kalluri R, Weinberg RA. The basics of epithelial‐mesenchymal transition. J Clin Invest. 2009;119:1420‐1428. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13. Uchikado Y, Natsugoe S, Okumura H, et al. Slug expression in the E‐cadherin preserved tumors is related to prognosis in patients with esophageal squamous cell carcinoma. Clin Cancer Res. 2005;11:1174‐1180. [PubMed] [Google Scholar]
  • 14. Javle MM, Gibbs JF, Iwata KK, et al. Epithelial‐mesenchymal transition (EMT) and activated extracellular signal‐regulated kinase (p‐Erk) in surgically resected pancreatic cancer. Ann Surg Oncol. 2007;14:3527‐3533. [DOI] [PubMed] [Google Scholar]
  • 15. Soltermann A, Tischler V, Arbogast S, et al. Prognostic significance of epithelial‐mesenchymal and mesenchymal‐epithelial transition protein expression in non‐small cell lung cancer. Clin Cancer Res. 2008;14:7430‐7437. [DOI] [PubMed] [Google Scholar]
  • 16. Guinney J, Dienstmann R, Wang X, et al. The consensus molecular subtypes of colorectal cancer. Nat Med. 2015;21:1350‐1356. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17. Hillman EM, Amoozegar CB, Wang T, et al. In vivo optical imaging and dynamic contrast methods for biomedical research. Philos Trans A Math Phys Eng Sci. 2011;369:4620‐4643. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. Imamura T, Saitou T, Kawakami R. In vivo optical imaging of cancer cell function and tumor microenvironment. Cancer Sci. 2018;109:912‐918. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19. Chen C, Yue D, Lei L, et al. Promoter‐operating targeted expression of gene therapy in cancer: current stage and prospect. Mol Ther Nucleic Acids. 2018;11:508‐514. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20. Haddad D, Fong Y. Molecular imaging of oncolytic viral therapy. Mol Ther Oncolytics. 2015;1:14007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21. Morin GB. The human telomere terminal transferase enzyme is a ribonucleoprotein that synthesizes TTAGGG repeats. Cell. 1989;59:521‐529. [DOI] [PubMed] [Google Scholar]
  • 22. Nakamura TM, Morin GB, Chapman KB, et al. Telomerase catalytic subunit homologs from fission yeast and human. Science. 1997;277:955‐959. [DOI] [PubMed] [Google Scholar]
  • 23. Harrington L, McPhail T, Mar V, et al. A mammalian telomerase‐associated protein. Science. 1997;275:973‐977. [DOI] [PubMed] [Google Scholar]
  • 24. Feng J, Funk WD, Wang SS, et al. The RNA component of human telomerase. Science. 1995;269:1236‐1241. [DOI] [PubMed] [Google Scholar]
  • 25. Nakayama J, Tahara H, Tahara E, et al. Telomerase activation by hTRT in human normal fibroblasts and hepatocellular carcinomas. Nat Genet. 1998;18:65‐68. [DOI] [PubMed] [Google Scholar]
  • 26. Takakura M, Kyo S, Kanaya T, Tanaka M, Inoue M. Expression of human telomerase subunits and correlation with telomerase activity in cervical cancer. Cancer Res. 1998;58:1558‐1561. [PubMed] [Google Scholar]
  • 27. Kishimoto H, Kojima T, Watanabe Y, et al. In vivo imaging of lymph node metastasis with telomerase‐specific replication‐selective adenovirus. Nat Med. 2006;12:1213‐1219. [DOI] [PubMed] [Google Scholar]
  • 28. Kojima T, Hashimoto Y, Watanabe Y, et al. A simple biological imaging system for detecting viable human circulating tumor cells. J Clin Invest. 2009;119:3172‐3181. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29. Shigeyasu K, Tazawa H, Hashimoto Y, et al. Fluorescence virus‐guided capturing system of human colorectal circulating tumour cells for non‐invasive companion diagnostics. Gut. 2015;64:627‐635. [DOI] [PubMed] [Google Scholar]
  • 30. Watanabe M, Kagawa S, Kuwada K, et al. Integrated fluorescent cytology with nano‐biologics in peritoneally disseminated gastric cancer. Cancer Sci. 2018;109:3263‐3271. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31. Yabusaki M, Sato J, Kohyama A, et al. Detection and preliminary evaluation of circulating tumor cells in the peripheral blood of patients with eight types of cancer using a telomerase‐specific adenovirus. Oncol Rep. 2014;32:1772‐1778. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. Sakurai F, Narii N, Tomita K, et al. Efficient detection of human circulating tumor cells without significant production of false‐positive cells by a novel conditionally replicating adenovirus. Mol Ther Methods Clin Dev. 2016;3:16001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33. Chen CZ, Li L, Lodish HF, Bartel DP. MicroRNAs modulate hematopoietic lineage differentiation. Science. 2004;303:83‐86. [DOI] [PubMed] [Google Scholar]
  • 34. Merkerova M, Belickova M, Bruchova H. Differential expression of microRNAs in hematopoietic cell lineages. Eur J Haematol. 2008;81:304‐310. [DOI] [PubMed] [Google Scholar]
  • 35. Bergelson JM, Cunningham JA, Droguett G, et al. Isolation of a common receptor for Coxsackie B viruses and adenoviruses 2 and 5. Science. 1997;275:1320‐1323. [DOI] [PubMed] [Google Scholar]
  • 36. Togo S, Katagiri N, Namba Y, et al. Sensitive detection of viable circulating tumor cells using a novel conditionally telomerase‐selective replicating adenovirus in non‐small cell lung cancer patients. Oncotarget. 2017;8:34884‐34895. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37. Zhang W, Ge K, Zhao Q, et al. A novel oHSV‐1 targeting telomerase reverse transcriptase‐positive cancer cells via tumor‐specific promoters regulating the expression of ICP4. Oncotarget. 2015;6:20345‐20355. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38. Zhang W, Bao L, Yang S, et al. Tumor‐selective replication herpes simplex virus‐based technology significantly improves clinical detection and prognostication of viable circulating tumor cells. Oncotarget. 2016;7:39768‐39783. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39. Kyo S, Takakura M, Fujiwara T, Inoue M. Understanding and exploiting hTERT promoter regulation for diagnosis and treatment of human cancers. Cancer Sci. 2008;99:1528‐1538. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40. Hashimoto Y, Watanabe Y, Shirakiya Y, et al. Establishment of biological and pharmacokinetic assays of telomerase‐specific replication‐selective adenovirus. Cancer Sci. 2008;99:385‐390. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41. Fujiwara T, Kagawa S, Kishimoto H, et al. Enhanced antitumor efficacy of telomerase‐selective oncolytic adenoviral agent OBP‐401 with docetaxel: preclinical evaluation of chemovirotherapy. Int J Cancer. 2006;119:432‐440. [DOI] [PubMed] [Google Scholar]
  • 42. Yano S, Takehara K, Kishimoto H, et al. OBP‐401‐GFP telomerase‐dependent adenovirus illuminates and kills high‐metastatic more effectively than low‐metastatic triple‐negative breast cancer in vitro. Cancer Gene Ther. 2017;24:45‐47. [DOI] [PubMed] [Google Scholar]
  • 43. Ishikawa W, Kikuchi S, Ogawa T, et al. Boosting replication and penetration of oncolytic adenovirus by paclitaxel eradicate peritoneal metastasis of gastric cancer. Mol Ther Oncolytics. 2020;18:262‐271. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44. Castano AP, Mroz P, Hamblin MR. Photodynamic therapy and anti‐tumour immunity. Nat Rev Cancer. 2006;6:535‐545. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45. Agostinis P, Berg K, Cengel KA, et al. Photodynamic therapy of cancer: an update. CA Cancer J Clin. 2011;61:250‐281. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46. Takehara K, Tazawa H, Okada N, et al. Targeted photodynamic virotherapy armed with a genetically encoded photosensitizer. Mol Cancer Ther. 2016;15:199‐208. [DOI] [PubMed] [Google Scholar]
  • 47. Takehara K, Yano S, Tazawa H, et al. Eradication of melanoma in vitro and in vivo via targeting with a Killer‐Red‐containing telomerase‐dependent adenovirus. Cell Cycle. 2017;16:1502‐1508. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48. Kamizono J, Nagano S, Murofushi Y, et al. Survivin‐responsive conditionally replicating adenovirus exhibits cancer‐specific and efficient viral replication. Cancer Res. 2005;65:5284‐5291. [DOI] [PubMed] [Google Scholar]
  • 49. Seo HK, Seo JB, Nam JK, et al. Development of replication‐competent adenovirus for bladder cancer by controlling adenovirus E1a and E4 gene expression with the survivin promoter. Oncotarget. 2014;5:5615‐5623. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50. Yamamoto Y, Hiraoka N, Goto N, et al. A targeting ligand enhances infectivity and cytotoxicity of an oncolytic adenovirus in human pancreatic cancer tissues. J Control Release. 2014;192:284‐293. [DOI] [PubMed] [Google Scholar]
  • 51. Nishimoto T, Yoshida K, Miura Y, et al. Oncolytic virus therapy for pancreatic cancer using the adenovirus library displaying random peptides on the fiber knob. Gene Ther. 2009;16:669‐680. [DOI] [PubMed] [Google Scholar]
  • 52. Li X, Mao Q, Wang D, Xia H. A novel Ad5/11 chimeric oncolytic adenovirus for improved glioma therapy. Int J Oncol. 2012;41:2159‐2165. [DOI] [PubMed] [Google Scholar]
  • 53. Roschger C, Cabrele C. The Id‐protein family in developmental and cancer‐associated pathways. Cell Commun Signal. 2017;15:7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54. Lasorella A, Uo T, Iavarone A. Id proteins at the cross‐road of development and cancer. Oncogene. 2001;20:8326‐8333. [DOI] [PubMed] [Google Scholar]
  • 55. Warram JM, Borovjagin AV, Zinn KR. A genetic strategy for combined screening and localized imaging of breast cancer. Mol Imaging Biol. 2011;13:452‐461. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56. Richter JR, Mahoney M, Warram JM, Samuel S, Zinn KR. A dual‐reporter, diagnostic vector for prostate cancer detection and tumor imaging. Gene Ther. 2014;21:897‐902. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57. Lee SJ, Kim HS, Yu R, et al. Novel prostate‐specific promoter derived from PSA and PSMA enhancers. Mol Ther. 2002;6:415‐421. [DOI] [PubMed] [Google Scholar]
  • 58. Hwang JE, Joung JY, Shin SP, et al. Ad5/35E1aPSESE4: a novel approach to marking circulating prostate tumor cells with a replication competent adenovirus controlled by PSA/PSMA transcription regulatory elements. Cancer Lett. 2016;372:57‐64. [DOI] [PubMed] [Google Scholar]
  • 59. Siegel RL, Miller KD, Fuchs HE, Jemal A. Cancer statistics, 2021. CA Cancer J Clin. 2021;71:7‐33. [DOI] [PubMed] [Google Scholar]
  • 60. Brand RE, Nolen BM, Zeh HJ, et al. Serum biomarker panels for the detection of pancreatic cancer. Clin Cancer Res. 2011;17:805‐816. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61. Xu C, Sun Y, Wang Y, et al. CEA promoter‐regulated oncolytic adenovirus‐mediated Hsp70 expression in immune gene therapy for pancreatic cancer. Cancer Lett. 2012;319:154‐163. [DOI] [PubMed] [Google Scholar]
  • 62. Villanueva A. Hepatocellular carcinoma. N Engl J Med. 2019;380:1450‐1462. [DOI] [PubMed] [Google Scholar]
  • 63. Zhang J, Chen G, Zhang P, et al. The threshold of alpha‐fetoprotein (AFP) for the diagnosis of hepatocellular carcinoma: a systematic review and meta‐analysis. PLoS One. 2020;15:e0228857. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64. Peng SY, Chen WJ, Lai PL, Jeng YM, Sheu JC, Hsu HC. High alpha‐fetoprotein level correlates with high stage, early recurrence and poor prognosis of hepatocellular carcinoma: significance of hepatitis virus infection, age, p53 and beta‐catenin mutations. Int J Cancer. 2004;112:44‐50. [DOI] [PubMed] [Google Scholar]
  • 65. Yoon AR, Hong J, Kim M, Yun CO. Hepatocellular carcinoma‐targeting oncolytic adenovirus overcomes hypoxic tumor microenvironment and effectively disperses through both central and peripheral tumor regions. Sci Rep. 2018;8:2233. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66. Brabletz S, Schuhwerk H, Brabletz T, Stemmler MP. Dynamic EMT: a multi‐tool for tumor progression. EMBO J. 2021;40:e108647. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67. Fischer KR, Durrans A, Lee S, et al. Epithelial‐to‐mesenchymal transition is not required for lung metastasis but contributes to chemoresistance. Nature. 2015;527:472‐476. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68. Zhao Z, Zhu X, Cui K, et al. In vivo visualization and characterization of epithelial‐mesenchymal transition in breast tumors. Cancer Res. 2016;76:2094‐2104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69. Bornes L, van Scheppingen RH, Beerling E, et al. Fsp1‐mediated lineage tracing fails to detect the majority of disseminating cells undergoing EMT. Cell Rep. 2019;29(9):2565‐2569.e3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70. Lourenco AR, Ban Y, Crowley MJ, et al. Differential contributions of pre‐ and post‐EMT tumor cells in breast cancer metastasis. Cancer Res. 2020;80:163‐169. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71. Jolly MK, Boareto M, Huang B, et al. Implications of the hybrid epithelial/mesenchymal phenotype in metastasis. Front Oncol. 2015;5:155. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72. Ieda T, Tazawa H, Okabayashi H, et al. Visualization of epithelial‐mesenchymal transition in an inflammatory microenvironment‐colorectal cancer network. Sci Rep. 2019;9:16378. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73. Wang W, Douglas D, Zhang J, et al. Live‐cell imaging and analysis reveal cell phenotypic transition dynamics inherently missing in snapshot data. Sci Adv. 2020;6:eaba9319. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74. Liu Y, Cao X. Characteristics and significance of the pre‐metastatic niche. Cancer Cell. 2016;30:668‐681. [DOI] [PubMed] [Google Scholar]
  • 75. Yano S, Takehara K, Kishimoto H, et al. Adenoviral targeting of malignant melanoma for fluorescence‐guided surgery prevents recurrence in orthotopic nude‐mouse models. Oncotarget. 2016;7:18558‐18572. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76. Yano S, Takehara K, Miwa S, et al. Fluorescence‐guided surgery of a highly‐metastatic variant of human triple‐negative breast cancer targeted with a cancer‐specific GFP adenovirus prevents recurrence. Oncotarget. 2016;7:75635‐75647. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77. Takahashi K, Tanabe R, Ehata S, et al. Visualization of the cancer cell cycle by tissue‐clearing technology using the Fucci reporter system. Cancer Sci. 2021;112:3796‐3809. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78. Zhao N, Powell RT, Yuan X, et al. Morphological screening of mesenchymal mammary tumor organoids to identify drugs that reverse epithelial‐mesenchymal transition. Nat Commun. 2021;12:4262. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Cancer Science are provided here courtesy of Wiley

RESOURCES