Waiting
登录处理中...

Trial ends in Request Full Access Tell Your Colleague About Jove

Cancer Research

Culture and Imaging of Ex Vivo Organotypic Pseudomyxoma Peritonei Tumor Slices from Resected Human Tumor Specimens

Published: December 9, 2022 doi: 10.3791/64620

Summary

We describe a protocol for the production, culture, and visualization of human cancers, which have metastasized to the peritoneal surfaces. Resected tumor specimens are cut using a vibratome and cultured on permeable inserts for increased oxygenation and viability, followed by imaging and downstream analyses using confocal microscopy and flow cytometry.

Abstract

Pseudomyxoma peritonei (PMP) is a rare condition that results from the dissemination of a mucinous primary tumor and the resultant accumulation of mucin-secreting tumor cells in the peritoneal cavity. PMP can arise from various types of cancers, including appendiceal, ovarian, and colorectal, though appendiceal neoplasms are by far the most common etiology. PMP is challenging to study due to its (1) rarity, (2) limited murine models, and (3) mucinous, acellular histology. The method presented here allows real-time visualization and interrogation of these tumor types using patient-derived ex vivo organotypic slices in a preparation where the tumor microenvironment (TME) remains intact. In this protocol, we first describe the preparation of tumor slices using a vibratome and subsequent long-term culture. Second, we describe confocal imaging of tumor slices and how to monitor functional readouts of viability, calcium imaging, and local proliferation. In short, slices are loaded with imaging dyes and are placed in an imaging chamber that can be mounted onto a confocal microscope. Time-lapse videos and confocal images are used to assess the initial viability and cellular functionality. This procedure also explores translational cellular movement, and paracrine signaling interactions in the TME. Lastly, we describe a dissociation protocol for tumor slices to be used for flow cytometry analysis. Quantitative flow cytometry analysis can be used for bench-to-bedside therapeutic testing to determine changes occurring within the immune landscape and epithelial cell content.

Introduction

Pseudomyxoma peritonei (PMP) is rare syndrome with an incidence rate of 1 per million people per year1. Most PMP cases are caused by metastases from appendiceal neoplasms. Given that mice do not have a human-like appendix, modeling this type of cancer remains extremely challenging. While the primary disease is often curable by surgical resection, treatment options for metastatic disease are limited. Therefore, the rationale for developing this novel organotypic slice model is to study the pathobiology of PMP. To date, there are no appendiceal organoid models that can be perpetually cultured; however, a recent model was shown to be useful for the pharmacological testing of therapeutic agents and immunotherapy2. As such, we have adapted an organotypic slice culture system, which has been used in other types of human cancers, such as brain, breast, pancreas, lung, ovarian, and others3,4,5,6.

In addition to appendiceal neoplasms, PMP occasionally results from other tumor types, including ovarian cancers7, and in rare circumstances, intraductal papillary mucinous neoplasms8 and colon cancer9. Additionally, these tumors tend to grow slowly, with poor engraft rates in patient-derived xenograft (PDX) models10,11. Given these challenges, there is an unmet need to develop models to study this disease to begin to understand the pathobiology of PMP, and how these cancer cells: are recruited to the peritoneal surfaces, proliferate, and escape immune surveillance.

While cut from the systemic vascular circulation, tumor slices do contain cellular and acellular components, including the extracellular matrix, stromal cells, immune cells, cancer cells, endothelial cells, and nerves. This semi-intact microenvironment allows for the functional investigation of these cell types, which is uniquely advantageous compared to 3D organoid cultures, which consist only of cancer cells12. While organotypic slice cultures are advantageous in some respects, they are also inherently a low-throughput-based approach, compared to 3D organoids, which can be expanded, and are suitable for multiplexed investigational therapeutic drug screening13,14,15. In the case of PMP, there have been no reports documenting reliable establishment and perpetual passaging of PMP-derived organoids16. This is likely due to the slow growing nature of PMP-derived tumor cells, as well as the low number of malignant epithelial cells found within these mucinous tumors. Given the need to develop models to study PMP, organotypic slices are uniquely suited to study this disease. We present a protocol for preparing, imaging, and analyzing PMP from human specimens.

Subscription Required. Please recommend JoVE to your librarian.

Protocol

The deidentification and acquisition of all tissues were performed under an IRB-approved protocol at the University of California, San Diego.

1. Preparation of human PMP tissues for tissue processing and culture

  1. Transport of tumor tissues and microdissection
    1. Prepare the transport and culture media: complete 10% (v/v) Dulbecco's Modified Eagle Media (DMEM), 10% FBS, 2 mM L-Glutamine, 1% Penicillin/Streptomycin (Pen Strep).
    2. Upon tissue arrival and according to an institutional IRB-approved protocol, transfer PMP tumor tissues into 35 dishes with a 6 cm diameter containing complete DMEM.
    3. Micro-dissect solid regions of the tumor from any liquefied mucin using a scalpel. While mucin nodules embedded within solid portions of the tissue can be cut, remove all liquified regions of the tumor. In addition to removing any liquified mucin, or highly mucinous tissue regions, remove any tissue that is difficult to cut with a scalpel. Cut tissue nodules into smaller pieces, roughly 1 cm3 in size.
      NOTE: Obtaining tumor nodules cleaned of mucin and dense ECM will be essential for successful cutting using a vibratome. As quality control, tumor tissue that arrives within the research laboratory is first cut by a pathologist, which is subsequently distributed by the UCSD biorepository. Tissue donors who undergo palliative debulking of the omental cake are optimal cases for this protocol given a high disease burden, which enables more slice production from the increased mass of tissue availability for research.
      CAUTION: Working with human tissues requires Biosafety Level 2 (BSL2) certification. Check with the institution for training on BSL2 procedures.
  2. Agarose preparation and tissue embedding
    1. Prepare a 5% solution of low melt agarose in PBS without FBS on the same day of tissue arrival. Pay close attention to the agarose solution as it tends to boil quickly.
      NOTE: Low melt agarose is essential for embedding tissue pieces. High melt agarose will solidify at higher temperatures, reducing the viability of tissue slices if embedded. Additionally, FBS in the dissolving solution will cause precipitates to form.
    2. Place the tissue pieces obtained in 6 cm dishes without liquid. Place no more than 2-3 pieces per 6 cm dish so that they can be removed as agarose cubes without disturbing or touching other tissues.
    3. Add the liquid 5% agarose solution to 6 cm dishes containing the 1 cm3 tumor specimens. Add enough agarose to just cover the specimen. Once added, place the embedded tissues in a 4 °C refrigerator for 30 min.
  3. Mounting tissue specimens on the vibratome
    1. Remove the solidified agar from the refrigerator and cut the agarose cubes slightly larger than the width of the tissue using a scalpel.
    2. Apply super glue to the vibratome stage and gently place the agarose cube onto the super glue. Allow 1-2 min for the glue to solidify. At this point, the agarose cube with tissue should be fixed to the stage.
    3. Fix the blade to the vibratome and set the desired thickness of the tissue section. For optimal downstream imaging using confocal microscopy, sections should be roughly 150 to 250 microns. Hit the Start button and continue to cycle through cutting of the agarose tissue block until the tissue is completely cut.
      NOTE: If the tissue is not cutting, or dislodges from the agarose cube, consider embedding the tissue in a higher concentration agarose and trim off any additional tissue pieces that may be too dense to cut or sticks to the vibratome blade.
  4. Culturing tissue slices on permeable inserts
    1. Prepare a permeable insert plate for culture by adding 2 mL of complete DMEM media above and 3 mL below the permeable membrane.
    2. Gently lift the tissue slices out of the cutting chamber of the vibratome using a thin paintbrush and place two to four slices into permeable culture dishes containing complete DMEM media. After 24 h, aspirate the media using a pipette and replace the media with fresh culture media.
    3. Culture the slices for up to 7 days at 37 °C/5% CO2, replacing the media every 24 h. At this time point, add controls for cell killing (bortezomib) and testable chemotherapies 5-fluoruracil (5-FU) to the culture media to perform pharmacological intervention.
    4. After 7 days of culturing, expect about a 15%-25% loss of viability compared to the day 0 time point (immediately after cutting) under non-drug treated conditions (complete DMEM). Measure the viability using live-dead viability dyes such as calcein AM (live) and propidium Iodide (dead).

2. Confocal imaging of living human tissue slices

NOTE: Once the organotypic tumor slices have been prepared, it is essential to determine the tissue viability to perform an efficient and optimized downstream analysis. Given that tumor specimens are 150-250 µm thick, confocal, or two-photon microscopy is highly recommended over wide-field microscopes for determining studies in situ. Flow cytometry can also be used for determining the viability and cellular populations (methods are below). However, the spatial resolution is lost during flow cytometry analysis, and viability is likely underestimated given the need to disassociate tumor slices by mechanical and chemical methods.

  1. Reagent preparation and experimental setup
    1. Place tumor slices for confocal imaging analysis in a single well of a 12-well dish containing PBS with 1% FBS. For calcium imaging experiments, instead of PBS, incubate the slices in extracellular calcium solution prepared as follows: 125 mM NaCl, 5.9 mM KCl, 2.56 mM CaCl2, 1 mM MgCl2, 25 mM HEPES, 0.1% BSA, pH 7.4, 3 mM glucose, sterile filtered. Prepare this solution ahead of time and store it for up to 1 month at 4 °C (see section 2.1.8).
    2. Following the concentrations and protocols recommended by the manufacturer, stain samples with imaging dyes (see Table of Materials) to determine the viability of tissue slices. Image for 10 min-1 h after adding the viability dye.
    3. After incubation, transfer the slices to an optically clear, glass bottom Petri dish containing PBS with 1% FBS to be used for imaging on a confocal imaging device.
      NOTE: For confocal imaging, the Nikon A1R Confocal platform was used.
    4. Open the confocal imaging software. Begin imaging at 10x and select the XYZ imaging mode. Then, configure the acquisition settings as follows.
      1. Turn on the following lasers: 488 nm and 561 nm. Adjust the gain to 100 with laser power at 1%. Adjust the laser power and gain as needed during image acquisition. Depending on the image quality desired, select 512 x 512 pixels or 1024 x 1024 pixels for higher resolution.
    5. Select Remove Interlock, and then select Scan to initiate imaging.
    6. For calcium imaging, incubate tissue slices with Fluo-4-AM for 1 h protected from light. Follow the manufacture's protocol for dissolving Fluo-4-AM in DMSO. After 1 h, wash slices 3x with extracellular calcium solution and follow the imaging protocol in steps 2.1.4-2.1.5.
    7. For calcium imaging experiments using Fluo-4-AM, select XYZT and unselect the 561 nm laser to increase acquisition speed. Additionally, use the resonance scanner to further increase the speed of imaging.

3. Disassociation of living slices for flow cytometry

NOTE: Disassociation of living tissue slices can be used for several downstream applications, including immunotyping, assessment of the viability, and interrogation of the changes to cell populations after pharmacological intervention. Steps should be taken to ensure that tissue quality and cell viability are maintained during the disassociation process.

  1. Cut a small piece from the end of a 1 mL pipette tip to widen the opening to allow vigorous mechanical dissociation by pipetting for 1 min.
  2. Incubate slices at 37 °C with rotation for 5-15 min in 1 mL of digestion buffer consisting of high glucose DMEM, 10% gentle collagenase/hyaluronidase (GCH), 10% FBS, and 10% DNaseI (1 mg/mL stock).
    NOTE: Batch-dependent changes of collagenase/hyaluronidase are often found.
  3. Perform vigorous disruption of the tissue using mechanical dissociation 2-3 times during incubation. Be sure to check digestion, by eye, every 5 min for evidence of complete digestion. If needed, stop the enzymatic digestion by proceeding to step 3.1.4.
  4. Once digested, pipette the slices and the digestion media on top of a 70 µm filter placed on top of a 50 mL conical tube. Pick larger pieces using sterile forceps or a pipette.
  5. Using the blunt back end of a plastic 5 mL syringe, mash any larger undissociated slices and further wash with 4 mL of PBS containing 2% FBS.
  6. Take the disassociated cell supernatant in a 50 mL conical tube and spin at 300 x g for 5-10 min. Remove the supernatant and wash the pellet with 1 mL of PBS containing 2% FBS.
  7. To prepare the sample for flow cytometry, add the blocking buffer containing 50 µL of human Fc-block and leave at room temperature for 15 min. Perform extracellular staining using the respective antibodies in 50 µL of PBS containing 2% FBS.
    ​NOTE: There are many flow cytometry systems. Once the sample is prepared for flow cytometry, refer to the manufacturer's protocol for operation of the device.

4. Pharmacological intervention using living tissue slices for viability and proliferation analysis

NOTE: Once the organotypic tumor slices have been prepared, interventional approaches can be performed by using several methods, including drug testing, siRNA, as well as viral infection of living tissue slices. Here we will discuss drug testing, as well as downstream functional readouts, which include viability analysis using local proliferation.

  1. Prepare 10 mL of 10% v/v complete DMEM with 10% FBS, 2 mM L-Glutamine, 1% Pen Strep.
  2. Aliquot 5 mL of complete media for control and treatment conditions. To the remaining 5 mL of media, add bortezomib (2 μM) to induce cell death in tumor slices as a positive control.
    NOTE: Bortezomib, at high concentrations, is a cytotoxic drug that induces cell death. The other cytotoxic drugs may be used as positive control for cell killing.
  3. Using two to four tissue slices, which have been plated onto the permeable dishes, add the media prepared in step 4.2 to the dish, as well as any additional combination therapies to be used for downstream viability LIVE/DEAD analysis using confocal microscopy as described in step 2.1.2, or use a commercial luminescent viability analysis using sequentially matched slices.
    NOTE: Sequential slice matching is essential for luminescent viability analysis, given that luminescent viability assay relies on cellular ATP content. As internal controls are lost during cell lysis, similar-sized slices are required since results are dependent on the starting number of viable cells (i.e., unbalanced slices will result in unbalanced results). If the user does not obtain sequential slices, luminescent viability analysis is not possible, and as such another viability assessment method will be required (flow cytometry or confocal imaging based live cell analysis).
  4. Leave tissue slices in culture containing complete DMEM for 2-5 days, changing media as well as bortezomib and 5-FU every other day.
  5. For luminescent viability analysis, add 500 µL of PBS into a 12-well dish and transfer the sequentially matched tissue slices to the PBS solution using a paintbrush or use the suction of a 1 mL pipette gently to transfer these.
  6. Remove the PBS and add the luminescent viability analysis solution that was prepared. See the instruction manual for preparation of the reagent.
  7. Add 500 µL of the luminescent viability solution per condition and incubate with a slow rotation on the shaker at room temperature for 30 min. Read luminescence using a luminescence plate reader.

Subscription Required. Please recommend JoVE to your librarian.

Representative Results

In short, human tumor specimens from PMP are obtained under an IRB-approved protocol. The tissue is prepared, micro-dissected, and solidified in an agarose mold to be cut using a vibratome (Figure 1A; Video 1). Once cut, tissue slices are placed and cultured on permeable insert membranes (Figure 1B), which can be utilized for imaging assays in situ, as well as for cellular and functional interrogation using flow cytometry, confocal imaging analysis, and cytotoxicity assays. A representative workflow of the culture and processing of human PMP tissue slices is shown in Figure 1C. Initial imaging analysis can be performed by light microscopy (Figure 2A-B). The staining of mucin in tissue slices were visualized using Periodic acid-Schiff (PAS) staining (Figure 2C) as well as mucin-specific antibodies (Figure 2D). To assess the viability, slices cut from different regions are incubated in calcein AM and propidium iodide to determine their health and viability (Figure 2E-F). Most cells (roughly 85%) are viable (calcein AM+; green), while some cells are dead (propidium iodide; red) when visualized by confocal microscopy after initial cutting. The extracellular matrix can be visualized using an incidence white light at an angular position of 180°. Tumors treated with chemotherapy prior to surgery often show more PI signal. The proliferation of tissue slices can be tracked using EdU added in the slice culture media. For detection, a commercial cell proliferation assay kit was used, and the assay was performed on fixed slices after 72 h of culture. Actively proliferating cells in culture can be measured in the FITC channel (green). The cell nuclei (DAPI) will overlap with cells that have proliferated (EdU+) during culture (Figure 2G). These results are quantified to determine the number of proliferating cells (DAPI+ nuclei) within the tumor slice (Figure 2H). The results shown here indicate that cultured slices are viable and proliferative during slice culture.

To identify immune cell functionality, Ca2+ imaging studies were performed on living tumor slices (Figure 3; Video 2). Using this approach direct and paracrine signaling mechanisms can be identified. To label local immune cells, in situ cytolabeling was performed by incubating slices with a CD11b-PE conjugated antibody along with Fluo-4-AM (Figure 3A). Pseudocolor scaled images showing intracellular Ca2+ levels better allow for visualizing differential levels of intracellular Ca2+ (Figure 3B). Spontaneous activity was tracked and time lapse images are shown for before (Figure 3C), during (Figure 3D), and after (Figure 3E) an intracellular Ca2+ response within the highlighted (white dotted circle box; Figure 3A,B) CD11b+ immune cell. The raw traces of Ca2+ responses in the CD11b immune cell (red) along with other responsive (black) and non-responsive cells (blue) are shown (Figure 3F). Quantification of the area under the curve of the raw traces shown from the responding immune cell (CD11b+; red Figure 3F) compared to the quantification of the non-responding cell (blue; Figure 3F) were quantified (Figure 3G). These results indicate that live cell functional tracking can be performed utilizing this living tissue slice platform.

Downstream analysis using flow cytometry can be used to immunotype as well as investigate changes in cellular profiles in response to therapeutic agents. Results may be due to direct signaling or indirect (paracrine) signaling. Representative results of immunotyping of a patient tumor sample with PMP are shown in Figure 4A. The tumor immuno-profile was quantified and the donor specimen 337 was found to have high levels of M2-macrophages (Figures 4B). These results indicate that utilizing PMP-derived tissue slices allow for researchers to interrogate the unique donor cellular landscape of the patient tumor samples. Further analysis and interrogation of the molecular, cellular, and genomic landscape is warranted to strategize a bench-to-bedside therapeutic approach that may be beneficial for patient outcomes.

Pharmacotyping and downstream analysis of tissue slices from PMP human tissues are a direct method to measure chemoresistance and drug sensitivity (Figure 5). The results show cell killing of tumor slices as confirmed by cytotoxicity analysis as well as TUNEL confocal imaging in response to the cytotoxic drug bortezomib (2 μM) and 5-fluorouracil (5-FU; 2 μM) (Figure 5A-C). As tumor killing responses are highly heterogeneous, it is important to stratify data on the basis of tumor grade and prior HIPEC, as repeated chemotherapy has been shown to be an ineffective method to ablate tumor cells in vivo and ex vivo17,18. In addition to TUNEL analysis, luminescent viability analysis was performed using sequentially matched slices (Figure 5D). It is essential to use sequentially matched tumor slices when performing cytotoxicity assays, as unmatched slices will lead to unreliable results.

Figure 1
Figure 1: Schematic illustration of pseudomyxoma peritonei ex vivo organotypic slice culture platform with representative tissue slices. A representative workflow for preparing organotypic slice cultures for immunotyping and to investigate functional properties of pseudomyxoma tumors of appendiceal origin. (A) The image of a live mounted tumor nodule from a pseudomyxoma peritonei (PMP) neoplasm. The tumor nodule is embedded in agarose, which is fixed to a vibratome using super glue. (B) The representative distribution of cutting tumor slices and arrangement in a trans-well culture dish. (C) The scheme representing workflow of slice culture and interrogation of tumor slices. Please click here to view a larger version of this figure.

Figure 2
Figure 2: Confocal imaging and functional analysis of living tissue slices. (A) Brightfield image 2x magnification of a representative tissue slice cut from a patient with pseudomyxoma peritonei of appendiceal origin. (B) A 10x magnification of the representative tissue slice shown in A. (C) PAS staining of a tissue from a patient with pseudomyxoma peritonei. Scale bar 1 mm. (D) Immunofluorescence imaging using MUC2 antibody (green), EPCAM (red), and DAPI (blue). Scale bar 50 µm. (E) Immunofluorescence imaging of using live (calcein AM; green) and dead (propidium iodide; red) dyes from donor 39. Scale bar 50 µm. (F) Quantification of viability analysis using three separate slices from donor 39 during initial tissue cutting. Error bars are the standard error of the mean. (G) Immunofluorescence imaging of proliferation using EdU (green). Cell nuclei are labeled with DAPI (blue). (H) Quantification of the number of EdU positive cells per 1,000 cells. N = 3 representative slices from donor 39. Error bars are the standard error of the mean. Please click here to view a larger version of this figure.

Figure 3
Figure 3: Live cell tracking using calcium imaging of living tissue slices. (A) In situ cytolabeling of a living tissue slice from donor 292. CD11b labeled macrophages shown in red. Calcium imaging dye Fluo-4-AM (green). Macrophage co-labeled with CD11b and Fluo-4-AM highlighted in white. (B) Pseudocolor scale of a living tissue slice (shown in A). Macrophage highlighted in white as in A. Scale bar showing 100 µm applies to A. Pseudocolor scale bar shown as mean gray values. (C-E) High magnification image of Fluo-4-AM before (C), during (D), and after (E) calcium activation. (F) Quantification of calcium levels shown by raw traces. The CD11b+ macrophage highlighted in A-E is highlighted in red. A non-responding cell is highlighted in blue. The other responding cell traces are shown in black. (G) Quantification of the normalized area under the curve shown between CD11b+ macrophage and a non-responding cell. Please click here to view a larger version of this figure.

Figure 4
Figure 4: Immunotyping of living human tumor slices from pseudomyxoma peritonei. (A) Representative scheme for performing flow cytometry characterization of immune cell profiles from pseudomyxoma tumors of appendiceal origin. (B) The percentage of immune cell subsets are color coded as well as the donor average bar plotted. The color markers above are provided for immune cell subsets within pie charts. Please click here to view a larger version of this figure.

Figure 5
Figure 5: Representative analysis for pharmacological intervention using organotypic slice culture. (A) Immunofluorescence of representative images from TUNEL analysis on untreated slices (left column), 5-FU (2 μM; middle column), or Bortezomib (2 μM; right column) treated slices. (B) Quantification of TUNEL immunofluorescence as a percentage of TUNEL positive DAPI+ cells from donor 339. (C) Quantification of TUNEL immunofluorescence as a percentage of TUNEL positive PanCK+ cells from donor 339. (D) Quantification of drug-treated slices using luminescent viability analysis from donor 339 pooled from 2-3 sequentially matched biological replicates. CTL denotes control. Concentration for control, 5-FU (2 μM), and Bortezomib (2 μM). Please click here to view a larger version of this figure.

Video 1: Cutting living human tissue slices from metastatic mucinous tumor nodules. Please click here to download this Video.

Video 2: Calcium imaging of living organotypic tumor slices from pseudomyxoma peritonei. Pseudocolor images of maximum projection from confocal images of a living tissue slice loaded with Fluo-4-AM at time point 00:00. Scale bar is 50 μm (bottom right). Please click here to download this Video.

Subscription Required. Please recommend JoVE to your librarian.

Discussion

This manuscript describes a technique that can be used to culture, interrogate, and analyze human pseudomyxoma peritonei (PMP) tumor specimens. We have utilized numerous downstream functional assays to interrogate the tumor immune microenvironment and a platform for bench-to-bedside testing.

While the method is highly efficient in our hands, it will require some practice to cut tumor specimens using a vibratome. Namely, we encountered problems that were due to highly mucinous samples, as well as samples that were improperly microdissected to remove uncuttable scaffolds (abdominal wall, ascites, various ECM proteins). As such, microdissection of the PMP tissue to remove excess mucin is a critical step of this protocol.We report an 80% success rate in patients with solid nodules as previously reported. High grade samples were technically less challenging to cut18. Moreover, the preparation of agarose (without FBS) and solidification of the tissue in agarose is also important for this protocol in order to produce tissue slices. Additionally, it will be essential for the researcher to be aware of information regarding tissue conditions and surgical parameters of the specimen (location of the metastasis, pre-treated samples), as these will help novel insights, as well as help to solve potential troubleshooting issues. Another critical step in this protocol is in the tumor cell functional analysis. Here, it is essential to use an epithelial marker (such as cytokeratin staining) in combination with a functional readout (apoptosis, proliferation) to assess efficacy. This is necessary in this particular type of tumor, because many PMP tumors have less than 10% of the cellular composition as tumor cells18. As such, flow cytometry, or confocal imaging is the preferred method of analysis. However, high throughput assays using luminescence-based imaging may be appropriate in tumors with higher tumor cell content, or if assessing the role of therapeutics on the TME.

Despite being a powerful tool, the organotypic slice culture has certain limitations. For example, a challenge of this technique is the cellular heterogeneity within tumors. To account for tumor heterogeneity, sequentially cut slices from similar tumor regions should be matched between groups for treatment and cell viability analysis. Cutting and preparing slices requires practice to microdissect uncuttable tumor scaffolds. These skills are required for successful cutting and optimization of tissue slices to limit experimental biases. Another limitation is tissue viability during slice culture. While it is possible to maintain brain or cardiac slices for months in cultures19,20, slice cultures from PMP have currently only been tested for roughly 1 week in culture18, which prevents long-term experimental investigation, including chronic drug treatment and genetic manipulation. Improvements in culture methods such as adding supplements and growth factors may be necessary to prolong the lifespan of PMP slice cultures.

Organotypic slice culture is a widely used technique to study various organs, including the brain, kidney, and liver. We have adopted this technique for use during PMP, providing us with an alternative model system that, to the best of our knowledge, is the first model to examine the PMP tumor microenvironment in a semi-intact preparation. In conjunction with 3D patient-derived organoids and explant cultures, this technique offers a robust and flexible platform to rapidly assess treatment efficacy and allow immediate translation of novel therapies from bench to bedside.

Subscription Required. Please recommend JoVE to your librarian.

Disclosures

The authors declare that they have no competing financial interests.

Acknowledgments

The authors would like to thank Kersi Pestonjamasp from the Moores Cancer Center imaging core facility for help with the microscopes UCSD Specialized Cancer Support Center P30 grant 2P30CA023100. This work was additionally supported by a JoVE publication grant (JRW), as well as generous gifts from the estate of Elisabeth and Ad Creemers, the Euske Family Foundation, the Gastrointestinal Cancer Research Fund, and the Peritoneal Metastasis Research Fund (AML).

Materials

Name Company Catalog Number Comments
1 M CaCl2 solution Sigma 21115
1 M HEPES solution Sigma H0887
1 M MgCl2 solution  Sigma M1028
100 micron filter ThermoFisher 22-363-549
22 x 40 glass coverslips Daiggerbrand G15972H
3 M KCl solution Sigma 60135
5 M NaCl solution Sigma S5150
ATPγS  Tocris  4080
Bovine Serum Albumin Sigma A2153
Calcein-AM  Invitrogen L3224
CD11b  Biolegend 101228
CD206  Biolegend 321140
CD3 Biolegend 555333
CD4  Biolegend 357410
CD45  Biolegend 304006
CD8  Biolegend 344721
CellTiter-Glo  Promega G9681
DMEM  Thermo Fisher 11965084
DPBS  Sigma Aldrich D8537
FBS, heat inactivated ThermoFisher 16140071
Fc-block  BD Biosciences 564220
Fluo-4 Thermo Fisher F14201
Gentle Collagenase/Hyaluronidase  Stem Cell 7912
Imaging Chamber Warner Instruments RC-26
Imaging Chamber Platform Warner Instruments PH-1
LD-Blue  Biolegend L23105
L-Glutamine 200 mM ThermoFisher 25030081
LIVE/DEAD imaging dyes Thermofisher R37601
Nikon Ti microscope  Nikon Includes: A1R hybrid confocal scanner including a high-resolution (4096x4096) scanner, LU4 four-laser AOTF unit with 405, 488, 561, and 647 lasers, Plan Apo 10 (NA 0.8), 20X (NA 0.9) dry objectives. 
Peristaltic pump  Isamtec ISM832C
Propidium Iodide Invitrogen L3224
Vacuum silicone grease Sigma Z273554-1EA

DOWNLOAD MATERIALS LIST

References

  1. Bevan, K. E., Mohamed, F., Moran, B. J. Pseudomyxoma peritonei. World Journal of Gastrointestinal Oncology. 2 (1), 44-50 (2010).
  2. Votanopoulos, K. I., et al. Appendiceal cancer patient-specific tumor organoid model for predicting chemotherapy efficacy prior to initiation of treatment: A feasibility study. Annals of Surgical Oncology. 26 (1), 139-147 (2019).
  3. Holliday, D. L., et al. The practicalities of using tissue slices as preclinical organotypic breast cancer models. Journal of Clinical Pathology. 66 (3), 253-255 (2013).
  4. Koerfer, J., et al. Organotypic slice cultures of human gastric and esophagogastric junction cancer. Cancer Medicine. 5 (7), 1444-1453 (2016).
  5. Misra, S., et al. Ex vivo organotypic culture system of precision-cut slices of human pancreatic ductal adenocarcinoma. Scientific Reports. 9 (1), 2133 (2019).
  6. Ohnishi, T., Matsumura, H., Izumoto, S., Hiraga, S., Hayakawa, T. A novel model of glioma cell invasion using organotypic brain slice culture. Cancer Research. 58 (14), 2935-2940 (1998).
  7. Seidman, J. D., Elsayed, A. M., Sobin, L. H., Tavassoli, F. A. Association of mucinous tumors of the ovary and appendix. A clinicopathologic study of 25 cases. The Amerian Journal of Surgical Pathology. 17 (1), 22-34 (1993).
  8. Mizuta, Y., et al. Pseudomyxoma peritonei accompanied by intraductal papillary mucinous neoplasm of the pancreas. Pancreatology. 5 (4-5), 470-474 (2005).
  9. Gong, Y., Wang, X., Zhu, Z. Pseudomyxoma peritonei originating from transverse colon mucinous adenocarcinoma: A case report and literature review. Gastroenterology Research and Practice. 2020, 5826214 (2020).
  10. Fleten, K. G., et al. Experimental treatment of mucinous peritoneal metastases using patient-derived xenograft models. Translational Oncology. 13 (8), 100793 (2020).
  11. Kuracha, M. R., Thomas, P., Loggie, B. W., Govindarajan, V. Patient-derived xenograft mouse models of pseudomyxoma peritonei recapitulate the human inflammatory tumor microenvironment. Cancer Medicine. 5 (4), 711-719 (2016).
  12. Jiang, X., et al. Long-lived pancreatic ductal adenocarcinoma slice cultures enable precise study of the immune microenvironment. Oncoimmunology. 6 (7), 1333210 (2017).
  13. Sundstrom, L., Morrison, B., Bradley, M., Pringle, A. Organotypic cultures as tools for functional screening in the CNS. Drug Discovery Today. 10 (14), 993-1000 (2005).
  14. Liu, L., Yu, L., Li, Z., Li, W., Huang, W. Patient-derived organoid (PDO) platforms to facilitate clinical decision making. Journal of Translational Medicine. 19 (1), 40 (2021).
  15. Croft, C. L., Futch, H. S., Moore, B. D., Golde, T. E. Organotypic brain slice cultures to model neurodegenerative proteinopathies. Molecular Neurodegeneration. 14 (1), 45 (2019).
  16. Carr, N. J. New insights in the pathology of peritoneal surface malignancy. Journal of Gastrointestinal Oncology. 12, 216-229 (2021).
  17. Votanopoulos, K. I., et al. Outcomes of repeat cytoreductive surgery with hyperthermic intraperitoneal chemotherapy for the treatment of peritoneal surface malignancy. Journal of the American College of Surgeons. 215 (3), 412-417 (2012).
  18. Weitz, J., et al. An ex-vivo organotypic culture platform for functional interrogation of human appendiceal cancer reveals a prominent and heterogenous immunological landscape. Clinical Cancer Research. 28 (21), 4793-4806 (2022).
  19. Pitoulis, F. G., Watson, S. A., Perbellini, F., Terracciano, C. M. Myocardial slices come to age: an intermediate complexity in vitro cardiac model for translational research. Cardiovascular Research. 116 (7), 1275-1287 (2020).
  20. Habeler, W., Peschanski, M., Monville, C. Organotypic heart slices for cell transplantation and physiological studies. Organogenesis. 5 (2), 62-66 (2009).

Tags

Ex Vivo Organotypic Tumor Slices Pseudomyxoma Peritonei Appendiceal Cancer Therapeutics Pathobiology Preclinical Setting Tumor Microenvironment Colorectal Cancer Ovarian Cancer Cell Interactions Metastasis Transport And Culture Media DMEM Mucin Scalpel Agarose Preparation Tissue Embedding
Culture and Imaging of <em>Ex Vivo</em> Organotypic Pseudomyxoma Peritonei Tumor Slices from Resected Human Tumor Specimens
Play Video
PDF DOI DOWNLOAD MATERIALS LIST

Cite this Article

Weitz, J., Montecillo Gulay, K. C.,More

Weitz, J., Montecillo Gulay, K. C., Hurtado de Mendoza, T., Tiriac, H., Baumgartner, J., Kelly, K., Veerapong, J., Lowy, A. M. Culture and Imaging of Ex Vivo Organotypic Pseudomyxoma Peritonei Tumor Slices from Resected Human Tumor Specimens. J. Vis. Exp. (190), e64620, doi:10.3791/64620 (2022).

Less
Copy Citation Download Citation Reprints and Permissions
View Video

Get cutting-edge science videos from JoVE sent straight to your inbox every month.

Waiting X
Simple Hit Counter