Combination Suicide Gene Delivery with an Adeno-Associated Virus

Jun 7, 2019 - Current treatment approaches for hepatocellular carcinoma (HCC) have a narrow therapeutic index and alternate modes of treatment are thu...
0 downloads 0 Views 6MB Size
Article Cite This: Bioconjugate Chem. 2019, 30, 1754−1762

pubs.acs.org/bc

Combination Suicide Gene Delivery with an Adeno-Associated Virus Vector Encoding Inducible Caspase‑9 and a Chemical Inducer of Dimerization Is Effective in a Xenotransplantation Model of Hepatocellular Carcinoma Nusrat Khan,† Sridhar Bammidi,† Sourav Chattopadhyay,† and Giridhara R. Jayandharan*,† †

Downloaded via UNIV OF CAMBRIDGE on August 20, 2019 at 01:07:36 (UTC). See https://pubs.acs.org/sharingguidelines for options on how to legitimately share published articles.

Department of Biological Sciences and Bioengineering, Indian Institute of Technology, Kanpur, Uttar Pradesh 20816, India S Supporting Information *

ABSTRACT: Current treatment approaches for hepatocellular carcinoma (HCC) have a narrow therapeutic index and alternate modes of treatment are thus required. We have utilized a gene delivery vector containing inducible caspase 9 (iCasp9) gene, which is a synthetic analogue based on the mammalian caspase 9 and fused to a human FK506 binding protein that allows its conditional dimerization to a synthetic, small molecule [chemical inducer of dimerization, AP20187] and results in target cell apoptosis. In our studies, we have tested these synthetic vectors based on an adeno-associated virus platform for their potential anti-tumorigenic effect in human HCC cells in vitro and in a HCC tumor model developed in nude mice. Our data demonstrates that the iCasp9-AP20187 bioconjugate is able to trigger terminal effectors of cellular apoptosis and presents a viable approach for the potential treatment of HCC.



INTRODUCTION Hepatocellular carcinoma (HCC) is a malignancy of the liver. About 600,000 new cases of HCC are reported annually, making it the fifth most common cause of all cancers affecting humans.1−3 The mortality of this condition is also very high; approximately 250,000 deaths occur annually due to HCC.3 Management of this condition requires a multidisciplinary approach and is primarily dependent on the patient’s characteristics such as the disease stage, performance status, and their hepatic functional reserve. Surgical resection is useful, as it simultaneously removes the tumor and the underlying cirrhosis, thereby reducing the risk of disease recurrence and a prospect of long-term survival.4,5 However, most patients have unresectable disease at presentation because of poor liver function.6−8 Sorafenib was the first agent associated with substantial increase in the overall survival of patients with locally advanced or metastatic HCC.9,10 While similar advances such as transcatheter arterial chemoembolization (TACE) have improved the overall survival to 35% of HCC patients (range, 16−61%) in the last two decades,11 the current treatment approaches remain suboptimal due to increasing drug resistance12−14 or their adverse side effects.14,15 Thus, there is a need to develop alternate modes of treatment in patients with HCC who are otherwise ineligible for the current standard of care. Gene therapy offers a promising platform as an alternate mode of therapy. Various strategies for cancer gene therapy © 2019 American Chemical Society

including gene-directed enzyme/pro-drug therapy, immunotherapy, inhibition of oncogenes, activation of tumor suppressor genes, and anti-angiogenic therapy have been employed.16,17 The most extensively used approach is by delivery of a suicide gene that encodes for an enzyme, which can convert an otherwise nontoxic pro-drug into a toxic product within the tumors. Among the available suicide gene prodrug systems, Herpes simplex virus thymidine kinase (HSV-TK) and its substrate, ganciclovir (GCV), is one of the most widely investigated in multiple tumor models at both preclinical and clinical levels.18−20 However, one of the significant limitations of the system is the immunogenicity conferred by the viral transgene and the high cell-cycle dependency of the activated drug that limits its cytotoxic activity to only the dividing cells.21 Recently, alternative strategies to overcome this limitation has been proposed. An inducible caspase 9 (iCasp9) gene, which is a synthetic analogue based on the mammalian caspase 9 fused to a human FK506 binding protein (FKBP) that allows conditional dimerization to a synthetic, bioinert small molecule [chemical inducer of dimerization (CID), AP20187], has been tested to switch-off genes during T cell therapy.22,23 iCasp9 is nonimmunogenic, since apart from a single amino acid substitution Received: April 22, 2019 Revised: May 17, 2019 Published: June 7, 2019 1754

DOI: 10.1021/acs.bioconjchem.9b00291 Bioconjugate Chem. 2019, 30, 1754−1762

Article

Bioconjugate Chemistry in the FKBP and a short linker peptide sequence, it is a selfprotein. Moreover, the suicide trigger is independent of the cell cycle phase and thus can be useful in a chronic and slowgrowing cancer such as HCC. Several studies have shown the efficacy and specificity of iCasp9 to their target cells.22,24−27 Among the available viral vectors, adeno-associated virus (AAV) are most suitable for liver directed cancer gene therapy, due to its nonpathogenic nature and its potential for long-term gene expression.28,29 Recombinant AAV serotype 2 is an ideal vector for suicide gene transfer in liver malignancies since it is naturally hepatotrophic.30−32 In the present study, we tested a bioconjugate suicide gene/chemical dimerizer (AAV-iCasp9/ AP20187) approach for the first time, in suitable in vitro and in vivo models of HCC.



RESULTS Construction and Validation of an AAV Based iCasp9 Vector. The iCasp9 gene was subcloned into an AAV backbone under the control of a chicken β-actin promoter as detailed in Figure S1. To ascertain if the plasmid encoding iCasp9 gene is functional and can trigger apoptosis in target cells, human cervical carcinoma (HeLa) cells were transfected with ∼0.5 μg of the plasmid, p.CBa-iCasp9. After 24 h, transfected cells were exposed to different concentrations of B/ B homodimerizer drug (AP20187). As seen in Figure S2, there was a decrease in cell viability with only ∼50−60% of cells surviving at 48 h time point after treatment with 1−20 nm of AP20187, when compared to the mock-treated cells. A dose of above 1 nm did not provide enhanced cytotoxicity, possibly due to a saturation of iCasp9 dimerizing effect of the drug. These data highlight that iCasp9 is an efficient suicide gene system, with its activity pronounced even at nanomolar concentrations of AP20187, that triggers significant apoptosis of the target cancer cells. Recombinant AAV-iCasp9 Vector and AP20187 Bioconjugate Induces Significant Cell Death in a Hepatocellular Carcinoma Model in Vitro. To test the utility of the iCasp9 vectors, we first packaged the iCasp9 transgene into AAV serotype 2 vectors. The vectors were quantified for their physical particle titers [vector genomes (vg)/mL] by a quantitative PCR method described earlier.33 To test the efficacy of these vectors and AP20187 bioconjugate, we utilized human hepatocellular carcinoma (Huh7) cells, which are known to be an aggressive tumor cell line and an excellent model system to study HCC as demonstrated earlier in multiple studies.34−37 To examine the cytotoxic effect of AAV2 iCasp9 system in vitro, Huh7 cells were infected with AAV vectors at a multiplicity of infection (MOI) of 5 × 104 vgs/cell. A day later, cells were treated with 10 nM of AP20187. Two days later, we measured the survival of vector treated cells using a luminescence based ATP assay kit according to the manufacturer’s protocol (CellTiter-Glo, Promega, Madison, WI, USA). As can be seen in Figure 1, the cell viability was reduced significantly to ∼50% (100 vs 50%, p < 0.05) in AAV2 iCasp9 treated cells in comparison to mocktreated cells. Huh7 cells that were treated with a Triton X-100 showed enhanced cytotoxicity, while treatment with only the dimerizer drug, AP20187 was nontoxic to the cells. This suggests that a dose of 5 × 104 AAV2-iCasp9 vectors per cell is sufficient to achieve cytotoxicity of the target Huh7 cells and that the functional dimerization of AP20187 is specific to iCasp9 enzyme.

Figure 1. In vitro cytotoxicity of AAV2 iCasp9 and dimerizer drug, AP20187: Huh7 cells were infected with AAV2 vectors encoding the iCasp9 gene, at a multiplicity of infection (MOI) of 5 × 104 vgs/cell. Twenty-four hours after vector transduction, the cells were treated with a chemical inducer of dimerization, AP20187 at a concentration of 10 nM. Forty-eight hours after drug treatment, cytotoxicity was assessed across all conditions by an ATP dependent luminescence assay according to the manufacturer’s instruction (CellTiter-Glo, Promega). The percent viability of Huh7 cells across different conditions is shown in comparison to compared to mock (PBS)treated cells. The data is representative of five independent experiments. Triton X100: positive control for cytotoxic agent; AP20187: vehicle and drug control. *P < 0.05 vs mock treated cells.

AAV-iCasp9 and AP20187 Combination Therapy Inhibits Tumor Growth in a Xenograft Model of HCC in Vivo. To further explore whether the cytotoxic effect of AAV2 iCasp9 vectors noted in vitro can be translated in vivo, we first developed xenotransplantation models of HCC in athymic mice as described in the Experimental Procedures section. The transplanted mice developed visible and palpable tumor nodules within their flanks after subcutaneous (s.c) administration of Huh7 cells. Once the tumors reached an average volume of 200 mm3 in ∼15 to 20 days, we initiated suicide gene transfer (Figure 2). Animals in the treatment group were administered with 5 × 1010 vgs of AAV2iCasp9 vectors intratumorally in a final volume of 200 μL. The control group received the same amount of PBS. We then administered AP20187 at two different doses (1 or 2 mg/kg body weight) intraperitoneally and 24 h after the suicide gene transfer, as described earlier.38 Tumor growth and its volume was measured every 3 days. The followup of animals was limited to 10 days after suicide gene transfer as the tumor burden in the control group reaches at this point typically is large, making the animal moribund and thus were sacrificed based on ethical considerations.36 The representative growth pattern and the volume of the tumors in the experimental mice that received a high dose of AP20187 (2 mg/kg body weight) are shown in Figure 3. Briefly, 15−20 days after administration of Huh7 cells, the average tumor size was 200 mm3 in all the animals (100−345 mm3, n = 12). After administration of AAV vectors containing the suicide gene followed by 2 mg/kg body weight of AP20187, tumor volume in both the treated and control group increased at a similar rate (286 vs 345 mm3, p = 0.14). However, tumor growth was delayed after 5 days in the AAV2iCasp9/AP20187 treated animals in comparison to control 1755

DOI: 10.1021/acs.bioconjchem.9b00291 Bioconjugate Chem. 2019, 30, 1754−1762

Article

Bioconjugate Chemistry

Figure 2. Experimental outline for therapeutic testing of AAV-iCasp9 vector and AP20187 dimerizer drug in a xenotransplantation model of hepatocellular carcinoma.

Figure 3. Tumor regression after suicide gene therapy in a xenotransplant model of HCC in vivo. Nude mice that developed Huh7 tumors (100− 150 mm3) were randomly distributed in two groups, receiving either PBS (mock) or the treatment group. Animals in the treatment group received AAV2 iCasp9-vectors at a dose of 5 × 1010 vgs/animal intratumorally on day 0, when their volume reached a baseline level of ∼200 mm3 after 15− 20 days. AP20187 treatment at a high dose (2 mg/kg body weight) was initiated on day 1 and continued until day 5 after vector administration. Tumor size was measured every 3 days. Relative tumor volume (mm3) is expressed as mean ± SD. The tumors of the animals that received suicide gene therapy demonstrated significant tumor growth suppression when compared to the mock group (P < 0.05, n = 5 per group). (B) Photographs of representative animals from both the groups and the enucleated tumors harvested at the end of the study. AAV2 iCasp9 treated tumors show large areas of tumor necrosis (marked by arrow in Figure 5) that correlate with the appearance of darkly staining patches on the external surface.

animals (1000 vs 2600 mm3, p < 0.05) (Figure 3A). Indeed, treatment with AAV2iCasp9/AP20187 inhibited the growth of xenografted tumors by ∼4.1-fold on day 6 after therapy initiation and ∼-3.5-fold by day 8 after therapy initiation. However, we also noticed an increased mortality in mice treated with 2 mg/kg body weight (50% in treatment vs 16% in control group). Interestingly, previous studies that have used AP20187 have not reported any mortality at a similar dose.24 Concurrently, in xenotransplanted mice that received a low dose (1 mg/kg body weight) of AP20187, we observed a similar regression in the tumor volume but not in the control animals (Figure 4). The rate of tumor inhibition was ∼3.6-fold on day 8 and ∼3.1-fold on day 10 after tumor initiation. The representative growth curve of the tumors after AAV-iCasp9/ AP20187 treatment is shown in Figure 4A. These data indicate that the rate of tumor attenuation was slightly reduced in the

low dose group (1 mg/kg body weight) as compared to animals that received a higher dose (2 mg/kg bodyweight) of AP20187 (Figure 3). However, there was no mortality seen in the low dose treatment group. Taken together, these data suggest that the combination of AAV2-iCasp9 vectors and a low dose AP20187 is well-tolerated and therapeutic in a murine model of HCC. Histological Analysis of HCC Tumors. Animals bearing the tumor from control or AAV treated groups were humanely euthanized and the subcutaneous tumors were harvested after 28−30 days of tumor initiation or 10 days after suicide gene transfer. For morphological characterization, hematoxylin and eosin staining was performed on the tissue sections from tumor samples. In case of sections from the control group, only a few apoptotic cells were seen (Figure 5). A significant number of apoptotic cells and infiltration of lymphocytes was observed 1756

DOI: 10.1021/acs.bioconjchem.9b00291 Bioconjugate Chem. 2019, 30, 1754−1762

Article

Bioconjugate Chemistry

Figure 4. Anti-tumorigenic effect of AAV-iCasp9 vectors and low dose of AP20187. (A) Xenotransplanted mice were either mock (PBS) injected or injected intratumorally with AAV2 iCasp9 vectors (5 × 1010 vgs/animal) when the average tumor volume reaches ∼200 mm3 after 15−20 days. A day later, animals in the treatment group received intraperitoneally a reduced dose of AP20187 (1 mg/kg body weight) for the next 5 days: (A) Animals from the AAV2 iCasp9 + AP20187 group demonstrated significant tumor regression as compared to mock injected animals (P ≤ 0.05, n = 8 per group) with no death observed in the treatment group. (B) Representative photographs of animal from experimental groups and their enucleated tumors.

Figure 5. Histological analysis of xenograft tumor tissue. Tumors were excised from experimental BALB/c nude mice after 8−10 days of treatment initiation. The tumor mass was rinsed with phosphate buffered saline (PBS), fixed in 10% buffered formalin for 24 h, embedded in paraffin, sliced, and sections were stained with hematoxylin and eosin. Microscopic observation of tumor tissue revealed pyknosis and apoptosis in animals that were administered with AAV2 iCasp9 vectors followed by AP20187 administration at high dose (2 mg/kg body weight) or low dose (1 mg/kg body weight) as compared to untreated animals (200× and 400× magnification).

to the tumor tissue harvested from the control group (Figure 6A). To further assess the extent of DNA damage, we performed quantification of TUNEL-positive cells using ImageJ software. Our data shown in Figure 6B revealed a significantly higher number of TUNEL-positive cells (238 ± 54 vs 20 ± 3, p < 0.05) in tumors that received suicide gene therapy in comparison to the control group.

across the breadth of the tumors that had been injected with AAV2 iCasp9 vectors followed by AP20187 treatment. Furthermore, the apoptotic cells in the treatment group, demonstrated well-defined features such as cell shrinkage, condensation, and pyknotic nuclei (marked by arrows, Figure 5). Tumor Regression after Suicide Gene Transfer Coincides with DNA Damage in the Tissue. In order to further characterize the tumor regression at the molecular level, in response to iCasp9/AP20187 treatment, a terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling (TUNEL, Roche, Basel, Switzerland) assay was performed. This test detects DNA strand break in cells undergoing apoptosis and has been used widely as a biomarker for diagnostic evaluation of apoptosis along with caspase-3.39,40 Our screening revealed that tumor tissue from the iCasp9/ AP20187 treated group were markedly positive for TUNEL staining (red color) signifying massive cell death as compared



DISCUSSION HCC develops in an environment of pre-existing liver dysfunction and pathology and is often associated with cirrhosis and poor liver function. Broad spectrum interventions such as ablation or sorafenib are highly effective in mitigating the tumor burden but also known to lead to increased liver or systemic toxicity.41,42 Thus, an ideal treatment strategy is one that targets only the HCC but avoids damage to normal tissue. Gene therapy using a suicide gene offers significant scope to develop such a platform, particularly with AAV based serotypes 1757

DOI: 10.1021/acs.bioconjchem.9b00291 Bioconjugate Chem. 2019, 30, 1754−1762

Article

Bioconjugate Chemistry

Figure 6. iCasp9-AP20187 induced apoptosis by DNA damage in Huh7 xenograft tumors. Athymic nu/nu mice bearing subcutaneous Huh7 tumors were sacrificed 8−10 days after vector and dimerizer drug treatment. Tumors were excised, fixed, and cryosectioned for TUNEL assay (A) Representative micrographs showing massive apoptotic cells within the tumors harvested from the treatment group (AAV2 iCasp9 + AP20187) as compared to control animals (Magnification 200×). (B) Quantification of data from (A) by ImageJ software, identified significant difference in the number of TUNEL positive cells between the treated and control groups (p ≤ 0.05).

cell-cycle dependency of the activated drug limits their cytotoxic effect to proliferating cells with some bystander effect improving the overall phenotypic outcome.50 However, HCC is a relatively slow growing cancer and thus these systems are only modestly effective.17,51 Other suicide genes that are cell cycle independent, such as nitroreductase or purine nucleoside phosphorylase, convert a relatively harmless prodrug (e.g., CB1954 and 6-methylpurine-2-deoxyriboside) to a highly toxic and membrane permeable mutagenic agent, underlying their narrow therapeutic index in multiple tumor models such as colon and prostate carcinoma.17,52,53 In an attempt to obviate these issues, we have utilized iCasp9 gene, which in its enzymatic form directly activates downstream components of the intrinsic apoptosis pathway, including the caspase 3 enzyme which is the terminal effector of apoptosis.27,54 The activation of this pro-apoptotic pathway is direct and independent of the cell-cycle stage.44 This was evident in our in vivo studies, where we observed an acute and significant tumor regression within 5 days of AAV-iCasp9 vector/AP20187 conjugate administration. We further observed that intratumoral injections of AAViCasp9 vectors followed by a low dose of AP20187 drug, reduced the tumor size by 3.6-fold. Similar observations have been made in several reports,24,55 where tumor growth in nude

such as AAV2 or AAV8 that have a high liver targeting ability.30−32,43 In the case of HCC, many studies have explored the use of HSV-TK/Ganciclovir prodrug system for gene therapy either in vitro or in an allograft or xenograft model.44−46 We have demonstrated for the first time, the utility of a recombinant AAV2 vector to deliver a modified gene component of the human intrinsic apoptotic pathway (caspase 9) in combination with a dimerizer AP20187 to induce cell death in HCC models in vitro and in vivo. We have also evaluated the safety and efficacy of the dimerizer drug at a lower dose (1 mg/kg body weight) which has been not reported until date. We explored the use of iCasp9-AP20187 combination for HCC suicide gene therapy because of this transgene’s potential advantages over other suicide gene systems. HSV-TK and cytosine deaminase (CD) require days to even weeks to achieve a maximum cytotoxic effect.18,20,28,47,48 The other limitation of such enzyme encoding genes, HSV-TK or CD is that they are of nonmammalian origin, either from Herpes simplex virus or of bacterial origin (E. coli), respectively, which is associated with systemic side effects in the host.49 Furthermore, both HSV-TK and CD and their prodrug combination by their mechanism of action require actively dividing cells to exert their anti-tumorigenic effect.22 This high 1758

DOI: 10.1021/acs.bioconjchem.9b00291 Bioconjugate Chem. 2019, 30, 1754−1762

Article

Bioconjugate Chemistry

(Cambridge, MA, USA). Lyophilized AP20187 was dissolved in 100% ethanol at a concentration of 1 mM and stored at −20 °C until further use. Construction of AAV iCasp9 Transgene. An inducible caspase9 (iCasp9) gene containing plasmid obtained from Addgene repository in a retroviral backbone (MSCV-F-del Casp9 IRES GFP) was cloned into AAV backbone by restriction enzyme cloning strategy. The iCasp9 gene flanking Age I and Hind III sites (1.3kb) at the 5′ and 3′ end, respectively, was in vitro synthesized (GenScript, Piscataway, NJ, USA). Further, the 1.3 kb gene fragment was subcloned into a pdsAAV-EGFP plasmid by replacing the EGFP cassette with the iCasp9 gene cassette, using Age I and Hind III enzymes. The resulting construct was then further verified by restriction digestion and DNA sequencing (Figure S1) Generation of Recombinant AAV. Recombinant AAV particles were produced by a helper virus free packaging method. Briefly, AAV-293 cells were first grown to 80% confluency in forty 15 cm2 dishes and then transfected with AAV2 rep/cap plasmid (p.AAVR2/C2), transgene (p.AAVCBa- iCasp9) and adenoviral helper plasmids (p.helper) at equimolar ratio using polyethylenimine (PEI, Polysciences, Warrington, PA) as previously described.62 Cells were harvested 68 h after transfection, lysed, and treated with benzonase (25 units/ml; Sigma-Aldrich, St.Louis, MI, USA). Further, the vectors were purified by iodixanol gradient ultracentrifugation (OptiPrep; Sigma-Aldrich) followed by column chromatography (HiTrap SP column; GE Healthcare Life Sciences, Chicago, IL, USA). Vectors were concentrated, using Amicon Ultra 10K centrifugal filters (Millipore, Burlington, MA, USA). Genomic titers of the vectors were determined by a quantitative PCR method.33 An average of six replicate samples were assayed to measure the physical particle titers of AAV and the data is expressed as vg/mL. Cytotoxicity Assay. To ascertain the cytotoxic effect of AAV-iCasp9 plasmid constructs, about 3 × 104 of HeLa cells were mock transfected (phosphate buffered saline, PBS) or transfected with 0.5 μg of the plasmid vectors using PEI (Polysciences) as the transfection agent. Twenty-four hours later, we performed a dose finding study, where HeLa cells were exposed to different concentrations of AP20187 (0.1−20 nm). Forty-eight hours after the drug treatment, cell viability was assessed using a luminescence based ATP assay kit according to the manufacturer’s protocol (CellTiter-Glo, Promega, Madison, WI, USA). Percentage cell viability was estimated by the formula, Percent cell viability = Normalized luminescence of sample/Normalized luminescence of control cells × 100. To further validate the recombinant AAV2 vector containing iCasp9 gene, ∼5 × 103 Huh7 cells per well were seeded in 96well plate and infected with AAV2-iCasp9 vectors at an multiplicity of infection (MOI) of 5 × 104 vgs/cell. Twentyfour hours later, cells were treated with 10 nM of AP20187. Two days after the drug treatment, cell viability was assessed using a luminescence based ATP assay kit as detailed above. Suicide Gene Transfer and AP20187 Dimerizer Combination Therapy into a Murine Model of Hepatocellular Carcinoma. We used athymic nude mice in BALB/C background for our studies (obtained from National Institute of Nutrition, Hyderabad, India). The animal experiments were approved by the IIT-Kanpur Institutional Animal Ethics committee. All animal experiments were performed in accordance with the relevant institutional and national

mice that received the treatment was inhibited but not completely eradicated. For example, in a breast cancer xenograft model tumor growth rate was retarded after 30 days compared to the control group, using an inducible HSVTk/GCV system.55 These observations underscore the fact that suicide gene transfer, in its present stage of development, is less likely to be used as a monotherapy as highlighted by other studies as well.19,20,28 To enhance the efficacy of this mode of treatment, an adjunct low-dose chemotherapy may be required, as it is less likely to trigger systemic side-effects during treatment of HCC. In addition, the use of alternate AAV serotypes such as AAV8, which is known to be highly efficient in targeting the hepatocytes,43,56 may further improve the anti-tumorigenic effect of this iCasp9 vector system. Our study has certain limitations. We have used a ubiquitous promoter system (chicken β-actin) for delivering the iCasp9 gene, which is known to be efficacious in a variety of preclinical and clinical settings.57,58 However, it may be desirable to direct iCasp9 expression under the control of liver specific promoter such as human apolipoprotein core hepatic control region element (LP1),59 human α-antitrypsin (hAAT)60 or the use of human α-fetoprotein (AFP)45 for focused expression of iCasp9 gene in the HCC tumors. Second, while the use of a xenotransplantation model in nude mice is necessary due to the lack of appropriate model systems of HCC, the immunocompromised nature of these animals may mask the effects of host immunity to the vectors or their transgene products. Thus, the use of natural immunocompetent model of HCC, such as orthotopic HCC model in a transgenic mice Stk4−/−Stk3F/−,61 may be necessary before this strategy is scaled up for potential clinical applications in patients with HCC.



CONCLUSIONS

Our study has highlighted the therapeutic efficacy of an inducible caspase-9 and AP20187 based suicide gene therapy in attenuating the disease progression in HCC. Further optimization of this system with modifications to the regulatory elements of the transgene construct or the vector capsid proteins to improve iCasp9 expression into HCC cells are some of the promising avenues for translating this molecular therapeutic into potential clinical applications.



EXPERIMENTAL PROCEDURES Cell Lines and Reagents. Human hepatocellular carcinoma (Huh7) cell line was a kind gift from Dr. Saumitra Das, Indian Institute of Science, Bangalore. Human cervical carcinoma cells (HeLa) was obtained from American Type Culture Collection (ATCC, Manassas, USA). AAV-293 cells were purchased from Stratagene (San Diego, CA, USA). All cell lines were maintained in growth medium comprising of Iscove’s Modified Dulbecco’s Medium (IMDM, Gibco, Life Technologies, Carlsbad, USA) supplemented with 10% fetal bovine serum, FBS (Gibco), 10 μg/mL each of Ciprofloxacin (HiMedia Laboratories, Mumbai, India) and Piperacillin (MP Biomedicals, Santa Ana, CA, USA) at 37 °C and 5% CO2. The cell lines used in the study were authenticated by short tandem repeat profiling to ascertain their identity and to check for any cellular cross-contamination or genetic drift associated with routine culturing. B/B homodimerizer drug (CID or AP20187) was purchased from ARIAD Pharmaceuticals 1759

DOI: 10.1021/acs.bioconjchem.9b00291 Bioconjugate Chem. 2019, 30, 1754−1762

Bioconjugate Chemistry



guidelines and regulations. For xenotransplantation, Huh7 cells in culture were trypsinized briefly, washed with IMDM containing 10% FBS, and harvested by centrifugation at 300g. The cell pellets were resuspended in serum free IMDM. Approximately, 5 × 106 cells were admixed with 25% Matrigel (Sigma-Aldrich) in a 0.2 mL suspension and injected subcutaneously to the flank region of adult nude mice (6−8 weeks old) to generate HCC tumors. When the tumors reached a size of 150−200 mm3, groups of transplanted mice were administered with ∼5 × 1010 vgs of AAV iCasp9 vectors intratumorally. Subsequently, vector injected animals were randomized to receive a low (1 mg/kg body weight) or high dose (2 mg/kg body weight) of AP20187 (CID). Overall, the drug was administered thrice at 48 h interval, and a day after AAV administration. Experimental mice were monitored daily for gross examination and their tumors measured every 3 days with Vernier calipers in two perpendicular diameters. Tumor volume was calculated as described previously63 with the formula 0.5 × L × W2, where L is the largest diameter (mm) and W is the shortest diameter (mm). Histological Analysis. Tumors from the AAV2- iCasp9/ AP20187 administered or control group were harvested and fixed in 10% buffered formalin and processed further by paraffin embedding and tissue sectioning (10 μm thickness) for microscopic examination. A gross and a microscopic examination following hematoxylin and eosin staining was performed by a pathologist as previously described.64 In Situ TUNEL Assay. Tumor samples excised from euthanized animals were washed briefly with PBS followed by their fixation in 10% buffered formalin. Samples were then cryopreserved by mounting in a OCT medium (SigmaAldrich). Frozen tissue was cut into 10 μm sections with a cryotome (Leica, Wetzlar, Germany). Sections were rinsed with PBS and fixed in 4% paraformaldehyde for 1 h at room temperature. An in situ TUNEL assay was performed according to the manufacturer’s instructions (Roche, Basel, Switzerland) to identify cells undergoing apoptosis. Sections were further washed and mounted with DAPI (Thermo Fischer, Waltham, Massachusetts, USA) and the images were acquired by Leica microscope (Leica DM5000 B). A minimum of three tissue sections per tumor were analyzed for quantification of TUNEL positive cells and the quantification performed by ImageJ analysis as previously described.65 Statistical Analysis. Data between the control and test groups was analyzed by either the paired one-tailed Student’s t test or by two way analysis of variance tests (ANOVA) as applicable using the SigmaPlot 11.0 (Systat Software, San Jose, CA, USA) or GraphPad Prism 7.0 (GraphPad Software, La Jolla, CA, USA). A p value < 0.05 was considered to be statistically significant.



Article

AUTHOR INFORMATION

Corresponding Author

*E-mail: [email protected]. Phone: +91 512 2594086. Fax: +91 512 2594010. ORCID

Nusrat Khan: 0000-0002-6676-7269 Sridhar Bammidi: 0000-0002-3189-6140 Sourav Chattopadhyay: 0000-0001-6966-0793 Giridhara R. Jayandharan: 0000-0001-9353-9053 Author Contributions

NK, SB, SC performed the experiments; GRJ conceived and designed the experiments and interpreted the data; NK and GRJ wrote the paper. The data sets generated during the study are available from the corresponding author on reasonable request. Notes

The authors declare no competing financial interest.



ACKNOWLEDGMENTS This work was supported through a Nanomission grant SR/ NM/NS-1084/2016, and in part through a Wellcome Trust DBT India Alliance Senior fellowship and an IIT-Kanpur Initiation grant to GRJ. We thank Dr. S Ganesh, IIT Kanpur for providing us the TUNEL assay reagents.



ABBREVIATIONS HCC, Hepatocellular carcinoma; iCasp9, inducible caspase 9; FKBP, FK506 binding protein; HSV-Tk, Herpes simplex virus thymidine kinase; GCV, Ganciclovir; AFP, α-fetoprotein; AAV, Adeno-associated virus; CID, Chemical inducer of dimerization



REFERENCES

(1) Acharya, S. K. (2014) Epidemiology of hepatocellular carcinoma in India. J. Clin. Exp. Hepatol. 4, S27−S33. (2) An, C., Choi, Y. A., Choi, D., Paik, Y. H., Ahn, S. H., Kim, M. J., Paik, S. W., Han, K. H., and Park, M. S. (2015) Growth rate of earlystage hepatocellular carcinoma in patients with chronic liver disease. Clin. Mol. Hepatol. 21, 279−286. (3) Kumar, A., Acharya, S. K., Singh, S. P., Saraswat, V. A., Arora, A., Duseja, A., Goenka, M. K., Jain, D., Kar, P., Kumar, M., et al. (2014) The indian national association for study of the liver (INASL) consensus on prevention, diagnosis and management of hepatocellular carcinoma in india: the puri recommendations. J. Clin. Exp. Hepatol. 4, S3−S26. (4) Cormier, J. N., Thomas, K. T., Chari, R. S., and Pinson, C. W. (2006) Management of hepatocellular carcinoma. J. Gastrointest. Surg. 10, 761−780. (5) Karaman, B., Battal, B., Sari, S., and Verim, S. (2014) Hepatocellular carcinoma review: current treatment, and evidencebased medicine. World J. Gastroenterol. 20, 18059−18060. (6) Llovet, J. M., Real, M. I., Montaña, X., Planas, R., Coll, S., Aponte, J., Ayuso, C., Sala, M., Muchart, J., Solà, R., Rodés, J., et al. (2002) Arterial embolisation or chemoembolisation versus symptomatic treatment in patients with unresectable hepatocellular carcinoma: a randomised controlled trial. Lancet 359, 1734−1739. (7) Lin, D.-Y., Liaw, Y.-F., Lee, T.-Y., and Lai, C.-M. (1988) Hepatic arterial embolization in patients with unresectable hepatocellular carcinomaa randomized controlled trial. Gastroenterology 94, 453− 456. (8) Buch, S. C., Kondragunta, V., Branch, R. A., and Carr, B. I. (2008) Gender-based outcomes differences in unresectable hepatocellular carcinoma. Hepatol. Int. 2, 95−101.

ASSOCIATED CONTENT

S Supporting Information *

The Supporting Information is available free of charge on the ACS Publications website at DOI: 10.1021/acs.bioconjchem.9b00291. Schematic of the cloning strategy for incorporation of iCasp9 gene into an inverted terminal repeat containing AAV vector (Figure S1); Validation of AAV-iCasp9 plasmid vectors in vitro (Figure S2) (PDF) 1760

DOI: 10.1021/acs.bioconjchem.9b00291 Bioconjugate Chem. 2019, 30, 1754−1762

Article

Bioconjugate Chemistry

(2005) An inducible caspase 9 safety switch for T-cell therapy. Blood 105, 4247−4254. (27) Rossignoli, F., Grisendi, G., Spano, C., Golinelli, G., Recchia, A., Rovesti, G., Orsi, G., Veronesi, E., Horwitz, E. M., and Dominici, M. (2019) Inducible Caspase9-mediated suicide gene for MSC-based cancer gene therapy. Cancer Gene Ther. 26, 11−16. (28) Kanazawa, T., Mizukami, H., Okada, T., Hanazono, Y., Kume, A., Nishino, H., Takeuchi, K., Kitamura, K., Ichimura, K., and Ozawa, K. (2003) Suicide gene therapy using AAV-HSVtk/ganciclovir in combination with irradiation results in regression of human head and neck cancer xenografts in nude mice. Gene Ther. 10, 51−58. (29) Monahan, P. E., Jooss, K., and Sands, M. S. (2002) Safety of adeno-associated virus gene therapy vectors: a current evaluation. Expert Opin. Drug Saf. 1, 79−91. (30) Murphy, S. L., Bhagwat, A., Edmonson, S., Zhou, S., and High, K. A. (2008) High-throughput screening and biophysical interrogation of hepatotropic AAV. Mol. Ther. 16, 1960−1967. (31) Dane, A. P., Wowro, S. J., Cunningham, S. C., and Alexander, I. E. (2013) Comparison of gene transfer to the murine liver following intraperitoneal and intraportal delivery of hepatotropic AAV pseudoserotypes. Gene Ther. 20, 460−464. (32) Dion, S., Bourgine, M., Godon, O., Levillayer, F., and Michel, M.-L. (2013) Adeno-Associated Virus-mediated gene transfer leads to persistent hepatitis b virus replication in mice expressing HLA-A2 and HLA-DR1 molecules. J. Virol. 87, 5554−5563. (33) Aurnhammer, C., Haase, M., Muether, N., Hausl, M., Rauschhuber, C., Huber, I., Nitschko, H., Busch, U., Sing, A., Ehrhardt, A., et al. (2012) Universal real-time PCR for the detection and quantification of adeno-associated virus serotype 2-derived inverted terminal repeat sequences. Hum. Gene Ther: Methods. 23, 18−28. (34) Guariniello, S., Di Bernardo, G., Colonna, G., Cammarota, M., Castello, G., and Costantini, S. (2015) Evaluation of the selenotranscriptome expression in two hepatocellular carcinoma cell lines. Anal. Cell. Pathol. 2015, 419561. (35) Zhuo, Z., Hu, J., Yang, X., Chen, M., Lei, X., Deng, L., Yao, N., Peng, Q., Chen, Z., Ye, W., et al. (2015) Ailanthone inhibits Huh7 cancer cell growth via cell cycle arrest and apoptosis in vitro and in vivo. Sci. Rep. 5, 16185. (36) Sápi, J., Kovács, L., Drexler, D. A., Kocsis, P., Gajári, D., and Sápi, Z. (2015) Tumor volume estimation and quasi-continuous administration for most effective bevacizumab therapy. PLoS One 10, No. e0142190. (37) Nwosu, Z. C., Battello, N., Rothley, M., Piorońska, W., Sitek, B., Ebert, M. P., Hofmann, U., Sleeman, J., Wölfl, S., Meyer, C., et al. (2018) Liver cancer cell lines distinctly mimic the metabolic gene expression pattern of the corresponding human tumours. J. Exp. Clin. Cancer Res. 37, 211. (38) Yagyu, S., Hoyos, V., Del Bufalo, F., and Brenner, M. K. (2015) An inducible caspase-9 suicide gene to improve the safety of therapy using human induced pluripotent stem cells. Mol. Ther. 23, 1475− 1485. (39) Singh, S. S., Mehedint, D. C., Ford, O. H., 3rd, Jeyaraj, D. A., Pop, E. A., Maygarden, S. J., Ivanova, A., Chandrasekhar, R., Wilding, G. E., and Mohler, J. L. (2009) Comparison of ACINUS, caspase-3, and TUNEL as apoptotic markers in determination of tumor growth rates of clinically localized prostate cancer using image analysis. Prostate 69, 1603−1610. (40) Duan, W. R., Garner, D. S., Williams, S. D., Funckes-Shippy, C. L., Spath, I. S., and Blomme, E. A. (2003) Comparison of immunohistochemistry for activated caspase-3 and cleaved cytokeratin 18 with the TUNEL method for quantification of apoptosis in histological sections of PC-3 subcutaneous xenografts. J. Pathol. 199, 221−228. (41) Lahat, E., Eshkenazy, R., Zendel, A., Zakai, B. B., Maor, M., Dreznik, Y., and Ariche, A. (2014) Complications after percutaneous ablation of liver tumors: a systematic review. Hepatobiliary Surg. Nutr. 3, 317−323.

(9) Cheng, A. L., Kang, Y. K., Chen, Z., Tsao, C. J., Qin, S., Kim, J. S., Luo, R., Feng, J., Ye, S., Yang, T. S., et al. (2009) Efficacy and safety of sorafenib in patients in the Asia-Pacific region with advanced hepatocellular carcinoma: a phase III randomised, double-blind, placebo-controlled trial. Lancet Oncol. 10, 25−34. (10) Forner, A., Llovet, J. M., and Bruix, J. (2012) Hepatocellular carcinoma. Lancet 379, 1245−1255. (11) Llovet, J. M., and Bruix, J. (2003) Systematic review of randomized trials for unresectable hepatocellular carcinoma: Chemoembolization improves survival. Hepatology 37, 429−442. (12) Nowak, A. K., Stockler, M. R., Chow, P. K., and Findlay, M. (2005) Use of tamoxifen in advanced-stage hepatocellular carcinoma. A systematic review. Cancer 103, 1408−1414. (13) Zhou, S. L., Zhou, Z. J., Hu, Z. Q., Huang, X. W., Wang, Z., Chen, E. B., Fan, J., Cao, Y., Dai, Z., and Zhou, J. (2016) TumorAssociated Neutrophils Recruit Macrophages and T-Regulatory Cells to Promote Progression of Hepatocellular Carcinoma and Resistance to Sorafenib. Gastroenterology 150, 1646−1658. (14) Galun, D., Srdic-Rajic, T., Bogdanovic, A., Loncar, Z., and Zuvela, M. (2017) Targeted therapy and personalized medicine in hepatocellular carcinoma: drug resistance, mechanisms, and treatment strategies. J. Hepatocell. Carcinoma. 4, 93−103. (15) Bruix, J., Takayama, T., Mazzaferro, V., Chau, G. Y., Yang, J., Kudo, M., Cai, J., Poon, R. T., Han, K. H., Tak, W. Y., et al. (2015) Adjuvant sorafenib for hepatocellular carcinoma after resection or ablation (STORM): a phase 3, randomised, double-blind, placebocontrolled trial. Lancet Oncol. 16, 1344−1354. (16) Zarogoulidis, P., Darwiche, K., Sakkas, A., Yarmus, L., Huang, H., Li, Q., Freitag, L., Zarogoulidis, K., and Malecki, M. (2013) Suicide gene therapy for cancer − current strategies. J. Genet. Syndr. Gene Ther. 4, 16849. (17) Rajab, T. K., Nelson, P., Keung, E. Z., and Conrad, C. J. (2013) Suicide gene therapy against cancer. J. Genet. Syndr. Gene Ther. 4, 1−8. (18) Li, L. Q., Shen, F., Xu, X. Y., Zhang, H., Yang, X. F., and Liu, W. G. (2013) Gene therapy with HSV1-sr39TK/GCV exhibits a stronger therapeutic efficacy than HSV1-TK/GCV in rat C6 glioma cells. Sci. World J. 2013, 951343. (19) Rainov, N. G. (2000) A phase III clinical evaluation of herpes simplex virus type 1 thymidine kinase and ganciclovir gene therapy as an adjuvant to surgical resection and radiation in adults with previously untreated glioblastoma multiforme. Hum. Gene Ther. 11, 2389−2401. (20) Nasu, Y., Saika, T., Ebara, S., Kusaka, N., Kaku, H., Abarzua, F., Manabe, D., Thompson, T. C., and Kumon, H. (2007) Suicide gene therapy with adenoviral delivery of HSV-tK gene for patients with local recurrence of prostate cancer after hormonal therapy. Mol. Ther. 15, 834−840. (21) Niculescu-Duvaz, I., Spooner, R., Marais, R., and Springer, C. J. (1998) Gene-directed enzyme prodrug therapy. Bioconjugate Chem. 9, 4−22. (22) Di Stasi, A., Tey, S. K., Dotti, G., Fujita, Y., Kennedy-Nasser, A., Martinez, C., Straathof, K., Liu, E., Durett, A. G., Grilley, B., et al. (2011) Inducible apoptosis as a safety switch for adoptive cell therapy. N. Engl. J. Med. 365, 1673−1683. (23) Straathof, K. C., Pule, M. A., Yotnda, P., Dotti, G., Vanin, E. F., Brenner, M. K., Heslop, H. E., Spencer, D. M., and Rooney, C. M. (2005) An inducible caspase 9 safety switch for T-cell therapy. Blood 105, 4247−4254. (24) Xie, X., Zhao, X., Liu, Y., Zhang, J., Matusik, R. J., Slawin, K. M., and Spencer, D. M. (2001) Adenovirus-mediated tissue-targeted expression of a caspase-9-based artificial death switch for the treatment of prostate cancer. Cancer Res. 61, 6795−6804. (25) Wu, C., Hong, S. G., Winkler, T., Spencer, D. M., Jares, A., Ichwan, B., Nicolae, A., Guo, V., Larochelle, A., and Dunbar, C. E. (2014) Development of an inducible caspase-9 safety switch for pluripotent stem cell-based therapies. Mol. Ther.–Methods Clin. Dev. 1, 14053. (26) Straathof, K. C., Pulè, M. A., Yotnda, P., Dotti, G., Vanin, E. F., Brenner, M. K., Heslop, H. E., Spencer, D. M., and Rooney, C. M. 1761

DOI: 10.1021/acs.bioconjchem.9b00291 Bioconjugate Chem. 2019, 30, 1754−1762

Article

Bioconjugate Chemistry (42) Verslype, C., Van Cutsem, E., Dicato, M., Arber, N., Berlin, J. D., Cunningham, D., De Gramont, A., Diaz-Rubio, E., Ducreux, M., Gruenberger, T., et al. (2009) The management of hepatocellular carcinoma. Current expert opinion and recommendations derived from the 10th World Congress on Gastrointestinal Cancer, Barcelona. Ann. Oncol. 20, vii1−vii6. (43) Tenney, R. M., Bell, C. L., and Wilson, J. M. (2014) AAV8 capsid variable regions at the two-fold symmetry axis contribute to high liver transduction by mediating nuclear entry and capsid uncoating. Virology 454, 227−236. (44) Su, H., Lu, R., Ding, R., and Kan, Y. W. (2000) Adenoassociated viral-mediated gene transfer to hepatoma: thymidine kinase/interleukin 2 is more effective in tumor killing in nonganciclovir (GCV)-treated than in GCV-treated animals. Mol. Ther. 1, 509−515. (45) Su, H., Chang, J. C., Xu, S. M., and Kan, Y. W. (1996) Selective killing of AFP-positive hepatocellular carcinoma cells by adenoassociated virus transfer of the herpes simplex virus thymidine kinase gene. Hum. Gene Ther. 7, 463−470. (46) Wang, Y., Huang, F., Cai, H., Zhong, S., Liu, X., and Tan, W. S. (2008) Potent antitumor effect of TRAIL mediated by a novel adenoassociated viral vector targeting to telomerase activity for human hepatocellular carcinoma. J. Gene Med. 10, 518−526. (47) Wang, Y. G., Huang, P. P., Zhang, R., Ma, B. Y., Zhou, X. M., and Sun, Y. F. (2016) Targeting adeno-associated virus and adenoviral gene therapy for hepatocellular carcinoma. World J. Gastroenterol. 22, 326−337. (48) Torimura, T., Ueno, T., Taniguchi, E., Masuda, H., Iwamoto, H., Nakamura, T., Inoue, K., Hashimoto, O., Abe, M., Koga, H., et al. (2012) Interaction of endothelial progenitor cells expressing cytosine deaminase in tumor tissues and 5-fluorocytosine administration suppresses growth of 5-fluorouracil-sensitive liver cancer in mice. Cancer Sci. 103, 542−548. (49) Fan, L., Freeman, K. W., Khan, T., Pham, E., and Spencer, D. M. (1999) Improved artificial death switches based on caspases and FADD. Hum. Gene Ther. 10, 2273−2285. (50) Kawashita, Y., Ohtsuru, A., Kaneda, Y., Nagayama, Y., Kawazoe, Y., Eguchi, S., Kuroda, H., Fujioka, H., Ito, M., Kanematsu, T., et al. (1999) Regression of hepatocellular carcinoma in vitro and in vivo by radiosensitizing suicide gene therapy under the inducible and spatial control of radiation. Hum. Gene Ther. 10, 1509−1519. (51) Lu, L. C., Hsu, C. H., Hsu, C., and Cheng, A. L. (2016) Tumor heterogeneity in hepatocellular carcinoma: facing the challenges. Liver Cancer 5, 128−138. (52) Spencer, D. M. (2000) Developments in suicide genes for preclinical and clinical applications. Curr. Opin. Mol. Ther. 2, 433− 440. (53) Prosser, G. A., Copp, J. N., Syddall, S. P., Williams, E. M., Smaill, J. B., Wilson, W. R., Patterson, A. V., and Ackerley, D. F. (2010) Discovery and evaluation of Escherichia coli nitroreductases that activate the anti-cancer prodrug CB1954. Biochem. Pharmacol. 79, 678−687. (54) Wang, C., and Youle, R. J. (2009) The role of mitochondria in apoptosis. Annu. Rev. Genet. 43, 95−118. (55) Li, Z. B., Zeng, Z. J., Chen, Q., Luo, S. Q., and Hu, W. X. (2006) Recombinant AAV-mediated HSVtk gene transfer with direct intratumoral injections and Tet-On regulation for implanted human breast cancer. BMC Cancer 6, 66. (56) Lisowski, L., Dane, A. P., Chu, K., Zhang, Y., Cunningham, S. C., Wilson, E. M., Nygaard, S., Grompe, M., Alexander, I. E., and Kay, M. A. (2014) Selection and evaluation of clinically relevant AAV variants in a xenograft liver model. Nature 506, 382−386. (57) Gray, S. J., Foti, S. B., Schwartz, J. W., Bachaboina, L., TaylorBlake, B., Coleman, J., Ehlers, M. D., Zylka, M. J., McCown, T. J., and Samulski, R. J. (2011) Optimizing promoters for recombinant adenoassociated virus-mediated gene expression in the peripheral and central nervous system using self-complementary vectors. Hum. Gene Ther. 22, 1143−1153.

(58) Kermany, D. S., Huu, V. A. N., Fu, X., Zhang, K., Valentim, C. C. S., Zhang, C. L., Zhu, J., Zhang, R., Duan, Y., and Sun, X. (2018) Clinical applications of retinal gene therapies. Precis. Clin. Med. 1, 5− 20. (59) Nathwani, A. C., Cochrane, M., McIntosh, J., Ng, C. Y. C., Zhou, J., Gray, J. T., and Davidoff, A. M. (2009) Enhancing transduction of the liver by adeno-associated viral vectors. Gene Ther. 16, 60−69. (60) Hafenrichter, D. G., Wu, X., Rettinger, S. D., Kennedy, S. C., Flye, M. W., and Ponder, K. P. (1994) Quantitative evaluation of liver-specific promoters from retroviral vectors after in vivo transduction of hepatocytes. Blood 84, 3394−3404. (61) Reiberger, T., Chen, Y., Ramjiawan, R. R., Hato, T., Fan, C., Samuel, R., Roberge, S., Huang, P., Lauwers, G. Y., Zhu, A. X., Bardeesy, N., Jain, R. K., and Duda, D. G. (2015) An orthotopic mouse model of hepatocellular carcinoma with underlying liver cirrhosis. Nat. Protoc. 10, 1264−1274. (62) Xiao, X., Li, J., and Samulski, R. J. (1998) Production of HighTiter Recombinant Adeno-Associated Virus Vectors in the Absence of Helper Adenovirus. J. Virol. 72, 2224−2232. (63) Tomayko, M. M., and Reynolds, C. P. (1989) Determination of subcutaneous tumor size in athymic (nude) mice. Cancer Chemother. Pharmacol. 24, 148−154. (64) Fischer, A. H., Jacobson, K. A., Rose, J., and Zeller, R. (2008) Hematoxylin and eosin staining of tissue and cell sections. CSH Protoc. pdb.prot4986 2008, pdb.prot4986. (65) Choi, H. K., Yessayan, D., Choi, H. J., Schellenberger, E., Bogdanov, A., Josephson, L., Weissleder, R., and Ntziachristos, V. (2005) Quantitative analysis of chemotherapeutic effects in tumors using in vivo staining and correlative histology. Cell. Oncol. 27, 183− 190.

1762

DOI: 10.1021/acs.bioconjchem.9b00291 Bioconjugate Chem. 2019, 30, 1754−1762