Waiting
로그인 처리 중...

Trial ends in Request Full Access Tell Your Colleague About Jove

Cancer Research

Implementation of Minimally Invasive Brain Tumor Resection in Rodents for High Viability Tissue Collection

Published: May 9, 2022 doi: 10.3791/64048

Summary

The present protocol describes a standardized resection of brain tumors in rodents through a minimally invasive approach with an integrated tissue preservation system. This technique has implications for accurately mirroring the standard of care in rodent and other animal models.

Abstract

The present protocol describes a standardized paradigm for rodent brain tumor resection and tissue preservation. In clinical practice, maximal tumor resection is the standard-of-care treatment for most brain tumors. However, most currently available preclinical brain tumor models either do not include resection, or utilize surgical resection models that are time-consuming and lead to significant postoperative morbidity, mortality, or experimental variability. In addition, performing resection in rodents can be daunting for several reasons, including a lack of clinically comparable surgical tools or protocols and the absence of an established platform for standardized tissue collection. This protocol highlights the use of a multi-functional, non-ablative resection device and an integrated tissue preservation system adapted from the clinical version of the device. The device applied in the present study combines tunable suction and a cylindrical blade at the aperture to precisely probe, cut, and suction tissue. The minimally invasive resection device performs its functions via the same burr hole used for the initial tumor implantation. This approach minimizes alterations to regional anatomy during biopsy or resection surgeries and reduces the risk of significant blood loss. These factors significantly reduced the operative time (<2 min/animal), improved postoperative animal survival, lower variability in experimental groups, and result in high viability of resected tissues and cells for future analyses. This process is facilitated by a blade speed of ~1,400 cycles/min, which allows the harvesting of tissues into a sterile closed system that can be filled with a physiologic solution of choice. Given the emerging importance of studying and accurately modeling the impact of surgery, preservation and rigorous comparative analysis of regionalized tumor resection specimens, and intra-cavity-delivered therapeutics, this unique protocol will expand opportunities to explore unanswered questions about perioperative management and therapeutic discovery for brain tumor patients.

Introduction

Glioblastoma (GBM) is the most common and aggressive primary brain tumor in adults. Despite recent advances in neurosurgery, targeted drug development, and radiation therapy, the 5-year survival rate for GBM patients is less than 5%, a statistic that has not significantly improved in over three decades1. Hence, there is a need for more effective treatment strategies.

To develop new therapies, it is becoming increasingly apparent that investigational protocols need to (1) utilize translatable preclinical models that accurately recapitulate the tumor heterogeneity and microenvironment, (2) mirror the standard therapeutic regimen used in patients with GBM, which currently includes surgery, radiotherapy, and chemotherapy, and (3) account for the difference between resected core and residual, invasive tumor tissues2,3,4,5. However, most of the currently available preclinical brain tumor models either do not implement surgical resection or utilize surgical resection models that are relatively time-consuming, leading to a significant amount of blood loss or lack standardization. Furthermore, performing resection of rodent brain tumors can be challenging due to a lack of clinically comparable surgical tools or protocols and the absence of an established platform6 for systematic tissue collection (Table 1).

The present protocol aims to describe a standardized paradigm for rodent brain tumor resection and tissue preservation using a multi-functional non-ablative minimally invasive resection system (MIRS) and an integrated tissue preservation system (TPS) (Figure 1). It is expected that this unique technique will provide a standardized platform that can be utilized in various studies in preclinical research for GBM and other types of brain tumor models. Researchers investigating therapeutic or diagnostic modalities for brain tumors can implement this protocol to achieve a standardized resection in their studies.

Subscription Required. Please recommend JoVE to your librarian.

Protocol

All animal studies were approved by the University of Maryland and the Johns Hopkins University Institutional Animal Care and Use Committee. C57BL/6 female mice, 6-8 weeks of age, were used for the present study. The mice were obtained from commercial sources (see Table of Materials). All Biosafety Level 2 (BSL-2) regulations were followed, including the usage of masks, gloves, and gowns.

1. Initial intracranial tumor implantation

  1. At the initial phase of the study, intracranially inject each mouse with 100,000 cells (GL261 murine glioma cell line) suspended in 4 µL of phosphate-buffered saline (1x PBS) to a depth of 2.5 mm following the previously published report7.
  2. Quantify the tumor signal in each mouse using the in vivo imaging system8 9 days following tumor implantation.
    NOTE: If needed, stratify the mice into two groups based on tumor burden. In the present study, the two groups were: (1) mice with relatively small tumor burden (mean bioluminescent signal = 5.5e+006 ± 0.1e+006 photons/s, n = 10) and (2) mice with relatively large tumor burden (mean bioluminescent signal = 1.69e+007 ± 0.2e+007 photons/s, n = 10), (p < 0.05, Mann-Whitney test)9.
  3. Divide each group into two comparable subgroups.
    NOTE: In this study, the two subgroups were: untreated mice (n = 5) and mice with tumors undergoing surgical resection using the MIRS (n = 5), (p > 0.05, Mann-Whitney test)9.
  4. Starting from the day of the resection, track the tumor progression using the in vivo imaging system at a frequency based on the tumor growth pattern.
    NOTE: For the GL261 cell line, track the tumor progression on the day of the resection and then every 3-6 days.

2. Tumor resection using MIRS

  1. Anesthetize the mouse using an isoflurane-O2 gas mixture in an induction chamber or intraperitoneal injection of xylazine/ketamine solution.
    1. If using the gas anesthetic, set the gas flow rate to 1.0 mL/min and the vaporizer to 2.0% for anesthesia induction, typically requiring 3-5 min in the chamber (see Table of Materials).
    2. If using the injectable anesthetic, anesthetize the mice by injecting 0.2 mL of anesthetic solution (80-100 mg/kg of ketamine and 10-12.5 mg/kg of xylazine, see Table of Materials) intraperitoneally.
  2. Assess the animal for adequate sedation by pinching the toe. Apply ophthalmic ointment to the eyes to avoid dryness of the cornea. Administer an analgesic (0.03-0.06 mg/kg buprenorphine subcutaneously) prior to the procedure.
  3. Place the mouse onto the stereotactic frame (see Table of Materials) once full sedation has been confirmed.
    NOTE: If using the gas anesthetic, place the nose of the mouse in a nose cone where it will continue to receive the isoflurane-O2 mixture during the procedure (1.5%).
  4. Remove the previous staple, then remove the hair either with a depilatory cream or by shaving. Disinfect the skin with alternating cycles of a chlorhexidine/betadine-based scrub and alcohol. Then, using a sterile scalpel, create a 1 cm longitudinal midline incision along the previous surgical scar.
  5. Attach the MIRS handpiece to the stereotactic arm through the stage adapter/handpiece holder for enhanced stability and precision.
  6. Set up the MIRS machine (see Table of Materials) using the following settings (Figure 1).
    1. Insert the power cord set on the rear panel into the power cord receptacle. Turn the power to the system on or off by toggling (1 = ON, 0 = OFF).
    2. Insert one end of the nitrogen hose (supplied with the console) into the male fitting on the console's rear panel. Rotate the connection nut clockwise to tighten the connection.
      NOTE: The opposite end of the hose needs to be connected to the nitrogen supply.
    3. Before attaching the hose to the nitrogen supply, confirm that the supply pressure does not exceed 100 psig, which is the input supply pressure recommended for the console.
      NOTE: The console will generate its own aspiration when activated by the foot pedal once the nitrogen has been supplied to the console.
    4. Secure the lid of the vacuum port and seal it to avoid any leakage in the vacuum system. Any leaks in the aspiration system will affect the performance of the MIRS console.
    5. If the aspiration is below 17 during setup or priming, check that the aspiration knob on the front of the console is set at the maximum level (100), ensure there is no leakage in the aspiration system, and confirm that the nitrogen input supply pressure is correct.
    6. To connect the foot pedal to the console, insert the gray foot pedal connector into its gray receptacle until it clicks and fits into position.
      NOTE: The foot pedal connector connects to the console in one orientation, and it is keyed.
    7. To connect the handpiece to the console, insert the blue handpiece connector into its blue receptacle until it clicks and fits into position.
      NOTE: The handpiece connector connects to the console in one orientation, and it is keyed.
    8. Prime each handpiece before using the system by aspirating sterile fluid from a small bowl into the aperture, through the tubing and handpiece, and then into the canister to ensure that the inside of the tubing and handpiece are lubricated to reduce the tissue occlusions.
    9. Prepare for aspiration alone or aspiration with cutting by selecting the appropriate mode on the console's front panel. Initiate using the foot pedal.
  7. Insert the 23 G MIRS cannula into the burr hole to a depth of 2.5 mm.
  8. Initiate the resection process by depressing the foot pedal connected to the cannula. Perform one full cycle (360°) or more of resection using the control knob in the handpiece.
    NOTE: The more cycles performed, the more volume of tumor tissue resected.
  9. Once the resection process is complete, withdraw the 23 G MIRS cannula from the burr hole and use 5 mL of 1x PBS to flush the tubing and dislodge any residual debris.
  10. Remove the mouse from the stereotactic frame and close the wound with a stapler or 4-0 suturing material (see Table of Materials).
  11. Place the mouse on a heating pad or under a warming light during recovery from anesthesia before returning it to its cage.
  12. After the experiment is complete, purge the cannula via flushing. Alternate with chilled media and air to "push" all of the resected tissue back to the collection canister. Remove the collection canister from the system and cap off with the provided cap.
  13. After completing step 2.12, place the distal tip of the cannula into 3% H2O2 and apply suction at 24-25 in Hg to fill the suction line back to the suction collection canister and let stand for 60-90 s. Flush with sterile media pulsing air and media intermittently.
  14. Monitor the mice for any neurological signs (abnormal erratic movements or seizures) following the procedure.
  15. Euthanize the mice with severe neurological impairments (become lethargic, have a gaunt appearance, hunched back, or have erratic movements).
    NOTE: For the present study, 200 mg/kg of a commercially available euthanasia solution (see Table of Materials) was used to euthanize each mouse.

3. Tissue collection via TPS

  1. Immerse the tumor sample in a tissue culture dish containing an RBC lysis medium (see Table of Materials) for 5 min at room temperature.
    NOTE: The tissue harvested from the MIRS into the TPS will be primarily in the form of single cells along with small chunks of tissue.
  2. Place a 70 µm filter (see Table of Materials) on a 50 mL conical tube and use a plunger of a 5 mL syringe to pass the tumor sample through the filter.
  3. With a transfer pipette, use the RPMI-1640 media to facilitate the passing of cells and any tissue mass through the filter.
  4. Centrifuge at 428 x g for 5 min at 4 °C. Discard the supernatant by a pipette.
  5. Resuspend each sample in 5 mL of prepared RPMI-1640 medium.
  6. To increase tissue viability, especially if large tissue chunks are visualized on initial impression, add the required volumes of the enzyme cocktail (containing DNAse I, collagenase IV, dispase, Papain, and EDTA, see Table of Materials) to each sample. Use the vortex to mix the solutions.
    NOTE: Step 3.6 is optional. The composition of the enzyme cocktail (for a total volume of 5 mL/sample): 300 µL of DNAse I Grade II, 150 µL of Collagenase/Dispase (cleaves fibronectin, collagenase IV, I, and nonpolar amino acids), 250 µL of Papain (nonspecific protease), and 6 µL of 0.5 M EDTA.
  7. Place the samples in a shaker incubator set at 200 rpm, 37 °C for 20 min.
  8. After 20 min, spin the samples at 428 x g for 5 min at 4 °C. Discard the supernatant.
  9. Filter single cells through a 70 µm cell strainer and spin down at 274 x g for 3 min at 4 °C. Conduct cell viability analysis10 with Trypan Blue and Hemocytometer (see Table of Materials).
    NOTE: Day 0 viability ranges from 30%-70% and increases significantly within 2-3 days.
  10. Proceed to Steps 4, 5, or 6, depending on the viability test needed.

4. Growing cells in adherent culture

  1. In a certified laminar flow hood, resuspend the pellet in serum-containing adherent medium (such as DMEM, 10% fetal bovine serum (FBS), and 1% penicillin/streptomycin (P/S) solution) and plate cells in an adherent cell flask.
  2. Maintain the cells in a controlled incubated environment (37 °C, 5% CO2).

5. Growing cells in suspension culture (neurospheres)

  1. In a certified laminar flow hood, resuspend the pellet in serum-free complete stem cell medium11 and plate in a suspension flask.
  2. Maintain the cells in a controlled incubated environment (37 °C, 5% CO2) for 2-3 days to allow neurosphere formation.
  3. After visualization of the neurospheres in the culture medium, use Trypsin-EDTA or Accutase (see Table of Materials) to obtain single-cell suspensions for passaging.
    NOTE: As long as special care is taken during harvest and appropriate media supporting neural stem cells is used, the stem cells in the harvested tissue must form neurospheres within a few days.

6. Preparing cells for reimplantation

  1. Resuspend the pellet at a concentration of 100,000 live cells per 4 µL of 1x PBS.
  2. Immediately inject into naïve mice using the intracranial tumor implantation method (step 1).

7. Histological analysis

  1. Immediately following resection, extract and fix the brains in 4% paraformaldehyde (PFA) for 24 h12.
  2. Transfer the brains to a 30% sucrose solution until they are saturated with sucrose (sunken down to the bottom of the container).
  3. Transfer the brains to a 70% ethanol solution.
  4. Perform paraffin block embedding, sectioning, and standard hematoxylin and eosin (H&E) staining following previously published report13.
    NOTE: The thickness of each section taken for staining was 10 µm.

Subscription Required. Please recommend JoVE to your librarian.

Representative Results

Surgical resection using the MIRS results in a significant decrease in the tumor burden
In the group with a smaller tumor burden, the mean baseline bioluminescent signal was 5.5e+006 photons/s ± 0.2e+006 in the subgroup that underwent resection. Following resection, the mean bioluminescent signal decreased to 3.09e+006 photons/s ± 0.3e+006, (p <0.0001, Mann-Whitney test)9 (Figure 2). The bioluminescent signal increased in the following few days until the mice were euthanized. Similarly, in the group with a larger tumor burden, the mean baseline bioluminescent signal was 1.68e+007 photons/s ± 0.1e+007 in the subgroup that underwent resection. Following resection, the mean bioluminescent signal decreased to 5.19e+006 photons/s ± 0.2e+006, (p <0.0001, Mann-Whitney test)9. The bioluminescent signal increased in the following few days until the mice were euthanized.

Resection using the MIRS can be adjusted for the desired volume of resection
In pre-resection imaging of syngeneic CT2A tumors, the tumor can be generally identified as a heterogeneous mass at the inoculation site with disrupted parenchymal architecture and peritumoral edema and hemorrhage indicated by heterogeneous areas of T2-weighted (T2w) hypo- and hyper-intensity. The needle track used for stereotactic tumor cell injection can be identified on T2w MRI scans14.

The resection cavity can be identified on post-resection T2w MRI scans as a large round hypointense area at the tumor inoculation site (Figure 3). The resection procedure did not cause significant blood loss or disruption of the surrounding brain architecture. In some cases, fluid accumulated in the resection cavity. As shown in Figure 4, the resection volume significantly increased from 9.4 mm3 for one rotation of the cutting aperture to 23.2 mm3 for two rotations (p = 0.0117), allowing for adjustment of volume resection to optimize for a known tumor burden.

Tumor resection using the MIRS leads to a 7-day prolongation in the median survival of tumor-bearing mice without inducing any neurological signs
As shown in Figure 5, there was a prolongation in the survival of mice that underwent surgical resection in both groups with small (6 days) and large (7 days) tumors. In the group with a smaller tumor burden, the median survival of the control subgroup was 16 days, while the median survival of the subgroup that underwent resection was 22 days (p = 0.0044). Similarly, in the group with a larger tumor burden, the median survival of the control subgroup was 12 days, while the median survival of the subgroup that underwent resection was 19 days (p = 0.0043). In addition, none of the mice undergoing resection using the MIRS showed any sign of neurological injury after the procedure. This indicates that the MIRS can achieve safe resection.

The resected tissue using the MIRS has high in vitro and in vivo viability
Cells extracted from the resected tissue were filtered, quantified, and resuspended in the appropriate medium (Figure 6) before conducting in vitro or in vivo viability experiments. To examine the in vitro viability of the cells and to confirm resection of the tumor and not normal brain parenchyma, cells were grown in suspension culture. Neurosphere formation, an indicator for tumor initiation potential, occurred with minimal tissue processing post-resection. This suggested that the MIRS platform harvested well-dissociated tissue and had minimal impact on the health and viability of the resected tissue. Samples taken straight from the TPS to light microscopy appeared to be primarily in the form of single cells along with the presence of a few small chunks of tissue. Day 0 viability ranged from 30%-70% (this represents the viability after passing the sample through a 70 µm filter and before plating). The resection device has a cutting aperture that can rotate 360° on the axis of the resection probe, allowing for the resection of concentric tissue volumes. On average, 2-3 million cells can be harvested with one 360° turn of the cutting aperture, approximately 7 million cells with two turns, and a total of 12-14 million cells can be obtained with three 360° turns of the cutting aperture. After plating in suspension flasks containing serum-free complete stem cell medium, neurospheres were visible by light microscopy on Day 2 and by the naked eye by Day 7 (Figure 7).

To examine in vivo viability, extracted cells were intracranially implanted into naive C57BL/6 mice (n = 8 mice, 100,000 live cells/mouse). Tumor growth was confirmed using the in vivo imaging system. The tumor signal continued to increase until all animals were euthanized by day 14 post tumor implantation (median survival of 11 days).

The representative H&E tissue section (Figure 3B) contains a clear, circular resection cavity with a rim of blood products (deep pink), inflammation, and residual tumor cells (dark purple cells). Macroscopically, the residual tumor cells are mesenchymal in nature and infiltrative, exhibiting extensive perivascular invasion and proliferation. Microscopically, marked nuclear atypia and mitotic figures were identified. Tumor-associated macrophages were also identified around areas of infarcted tissue and microhemorrhages. Tissue architecture in the surrounding brain parenchyma did not appear to be disturbed.

Figure 1
Figure 1: MIRS system setup. (A) Minimally invasive resection system (MIRS) for brain tumor in rodent models with an integrated tissue preservation system. (B) Front panel of MIRS console. 1 = System Power, 2 = Foot Pedal, 3 = Prime Button, 4 = Aspiration, 5 = Aspiration Level Control Dial, 6 = Aspiration Level Indicator, 7 = Handpiece, 8 = Cutter Enable Button. (C) Rear panel of MIRS console. 1 = Power Cord Receptacle, 2 = Circuit Breaker, 3 = Nitrogen Supply Input. Please click here to view a larger version of this figure.

Figure 2
Figure 2: Changes in average tumor burden in mice with untreated tumors versus mice undergoing resection of tumors by MIRS. (A) Mice with small baseline tumor burden and (B) mice with large baseline tumor burden (p < 0.0001, SD ≤0.3e+006 in all groups at each time point and too small to be displayed on graph). Animals either succumbed to tumor burden or were euthanized. Please click here to view a larger version of this figure.

Figure 3
Figure 3: MRI and H&E analysis. (A) Pre- and post-resection T2-weighted brain MRI images and (B) a representative H&E-stained coronal brain section showing a clear, circular resection cavity with a rim of blood products (deep pink), inflammation, and residual tumor cells (dark purple cells). MRI images and H&E sections were obtained immediately after resection. Please click here to view a larger version of this figure.

Figure 4
Figure 4: Determination of tumor resection volume. The average volumes calculated from MRI images of resection cavities created with one vs. two rotations of the cutting aperture, n = 4 per group. p = 0.01, two-tailed T-test. Please click here to view a larger version of this figure.

Figure 5
Figure 5: Kaplan-Meier curves of mice with untreated tumors versus mice undergoing resection of tumors by MIRS. (A) Mice with small baseline tumor burden. (B) Mice with large baseline tumor burden. Please click here to view a larger version of this figure.

Figure 6
Figure 6: Viability test of the resected tumor cells. (A) Representative light microscopy images of tissue harvested with MIRS on Day 0 at 20x. (B) Kaplan-Meier curve describing survival of mice after intracranial implantation of cells harvested from resected tissue. Please click here to view a larger version of this figure.

Figure 7
Figure 7: Formation of neurospheres. Representative light microscopy images of neurospheres generated from resected tissue on Day 2 post-resection at (A) 20x and (B) 10x, and on day 7 post-resection at (C) 20x and (D) 10x. Please click here to view a larger version of this figure.

Minimally Invasive Resection System Historical Resection
Operative time and skills Minimal operative time (<2 min for each animal). Minimal skills needed. Surgical time is not standardized and surgical experience with small animals and microsurgery is beneficial.
Blood loss Minimal Unpredictable
Standardized volume of resection Volume of resection determined/adjusted by the number of rotations of the resection tool Volume varies greatly among subjects

Table 1: Comparison between the minimally invasive resection system (MIRS) and historical surgical resection models.

Subscription Required. Please recommend JoVE to your librarian.

Discussion

Tumor resection is a cornerstone of neurosurgical oncology treatment plans for both low-grade and high-grade brain tumors. Cytoreduction and debulking of the tumor correlate with improved neurological function and overall survival in patients with brain tumors1,2,5,6. Although protocols for surgical resection have been previously described in rodent models, these protocols have suffered from several limitations that can confound the generated results and the overall translatability of the preclinical model. For instance, previously reported surgical resection protocols have included a craniotomy with 3-4 small burr holes to create a bone flap and then skillful distinction and differentiation of the tumor's coloration and consistency compared to normal brain tissue6. These protocols are time-consuming and require the research personnel to have advanced skills in utilizing the surgical microscope, handling surgical instruments, identifying the tumor borders concerning the surrounding normal brain tissue, and achieving adequate homeostasis. Such procedures often lead to significant blood loss and animal death. In addition, it can be challenging to achieve standardized resection with comparable volumes of tumor tissue resected across different animals in the same experiment. This continuity and uniformity becomes more challenging with multiple personnel performing the procedure.

In contrast, the MIRS protocol described here removes these limitations (Table 1). The present protocol requires a minimally invasive approach through the same burr hole used for initial tumor implantation. As detailed in the protocol section, the resection tool can be installed on a stereotactic frame that allows for accurate insertion of the cannula into the tumor cavity. The speed of the blade and the number of resection cycles performed can be easily adjusted through a handpiece used by the research personnel to control the volume of tumor resected in a standardized fashion without causing a significant amount of blood loss.

As noted in the results, the mice that underwent resection with the MIRS device demonstrated prolonged survival compared to the cohort of mice with unresected tumors. This, along with the data, shows that tumor burden was significantly reduced in the resected group compared to the unresected control group, indicating that the MIRS device effectively debulks the tumor with minimal disturbance to surrounding healthy brain tissues. Further, in the days following resection, the tumor burden progressed steadily due to the presence of a residual tumor and the infiltrative nature of the CT-2A tumor cell line. The MIRS model can thus replicate the debulking process of infiltrative tumor types, which can be followed with chemotherapeutics or radiation therapy to more closely mirror the treatment regimen in human brain tumor treatment protocols15,16,17.

As is the case with any suction device, clogging of the aspiration tubing can be one of the limitations of the MIRS system. This can occur due to the accumulation of dry tissue or blood during or after using the machine. To avoid this, immediately at the end of each session of resections, purging the handpiece cannula is recommended by flushing with chilled media alternating with air to "push" all of the resected tissue to the collection canister, followed by flushing with 3%  H2O2. This will capture all of the tissue acquired during the resection and ensure none remains in the cannula or tubing. Furthermore, if the system is not aspirating/cutting, this can be because the handpiece is not plugged in, the resection cannula cutter is not turned on, the aspiration line from the console is not connected to the canister, the canister lid is not tight, or the foot pedal is not plugged in.

In addition, while we have shown that the MIRS can be used for effective and standardized resection with high in vitro and in vivo viability of the resected tissue, future studies are encouraged to further investigate other facets of implementing such systems in brain tumor research. These include examining the impact of the resection process on the molecular and cellular components of the tumor mass and the tumor microenvironment. In addition, studies are needed to confirm that the resected tissue using the MIRS device recapitulates the parental tumor15.

In conclusion, a protocol of a minimally invasive approach is described for standardized surgical resection in a rodent brain tumor model that is coupled with an integrated and automated tissue preservation system. This protocol paves the path toward establishing highly translational and predictive preclinical brain tumor research models. Future applications of this protocol can potentially include preclinical studies investigating different therapeutic or diagnostic modalities for brain tumors which can implement this protocol to achieve a standardized resection.

Subscription Required. Please recommend JoVE to your librarian.

Disclosures

BT has research funding from NIH and is a co-owner for Accelerating Combination Therapies*, and Ashvattha Therapeutics Inc. has licensed one of her patents. GW has NIH funding (R01NS107813). HB is a paid consultant to Insightec and chairman of the company's Medical Advisory Board. This arrangement has been reviewed and approved by Johns Hopkins University following its conflict-of-interest policies. HB has research funding from NIH, Johns Hopkins University, and philanthropy and is a consultant for CraniUS, Candel Therepeutics, Inc., Accelerating Combination Therapies*, Catalio Nexus Fund II, LLC*, LikeMinds, Inc*, Galen Robotics, Inc.* and Nurami Medical*. (*includes equity or options).

Materials

Name Company Catalog Number Comments
1 mL syringes BD 309628
15 mL conical tubes Corning 430052
200 proof ethanol PharmCo 111000200
5 mL pipettes CoStar 4487
70 micron filter Fisher 08-771-2
Accutase Millipore Sigma SIG-SCR005
Anased (Xylazine injection, 100 mg/mL) Covetrus 33198
Anesthesia System Patterson Scientific 78935903
Anesthesic Gas Waste Container Patterson Scientific 78909457
Bench protector underpad Covidien 10328
C57Bl/6, 6-8 week old mice Charles River Laboratories Strain Code 027
ChroMini Pro Moser Type 1591-Q
Collagenase-Dispase Roche #10269638001
Countess II Automated Cell Counter Thermo Fisher
Countess II FL Hemacytometer Thermo Fisher A25750
Debris Removal Solution Miltenyi Biotech #130-109-398
D-Luciferin Goldbio LUCK-1G
DMEM F12 media Corning 10-090-CV
DMEM media Corning 10-013-CV
DNAse I Sigma Aldrich #10104159001
Eppendorf tubes Posi-Click 1149K01
Euthanasia solution Henry Schein 71073
FBS Millipore Sigma F4135
Fetal Bovine Serum Thermo Fisher 10437-028
Formalin Invitrogen INV-28906
Gauze Henry Schein 101-4336
hEGF PeproTech EC 100-15
Heparin Sigma H-3149
hFGF-b PeproTech EC 1001-18B
Induction Chamber Patterson Scientific 78933388
Isoflurane Covetrus 11695-6777-2
Isoflurane Vaporizer Patterson Scientific 78916954
Ketamine Covetrus 11695-0703-1
Kopf Stereotactic frame Kopf Instruments 5001
Lightfield Microscope BioTek Cytation 5
Microinjection Unit Kopf 5001
Micromotor drill Foredom F210418
MRI system Bruker 7T Biospec Avance III MRI Scanner
NICO Myriad System NICO Corporation
Ophthalmic ointment Puralube vet ointment
Papain Sigma Aldrich #P4762
PBS Invitrogen #14190250
PenStrep Millipore Sigma N1638
Percoll solution Sigma Aldrich  #P4937
Pipette controller Falcon A07260
Povidone-iodine solution Aplicare 52380-1905-08
Progesterone Sigma P-8783
Putrescine Sigma P-5780
RPMI Media Invitrogen INV-72400120
Scalpel blade Covetrus 7319
Scalpel handle Fine Science Tools 91003-12
Skin marker Time Out D538,851
Staple remover MikRon ACR9MM
Stapler MikRon ACA9MM
Staples Clay Adams 427631
Stereotactic Frame Kopf Instruments 5000
Sucrose Sigma Aldrich S9378
Suture, vicryl 4-0 Ethicon J494H
T-75 culture flask Sarstedt 83-3911-002
TheraPEAKTM ACK Lysing Buffer (1x) Lonza BP10-548E
Trypsin-EDTA Corning MDT-25-053-CI

DOWNLOAD MATERIALS LIST

References

  1. Mineo, J. F., et al. Prognosis factors of survival time in patients with glioblastoma multiforme: a multivariate analysis of 340 patients. Acta Neurochirurgica. 149 (3), 245-252 (2007).
  2. Miyai, M., et al. Current trends in mouse models of glioblastoma. Journal of Neuro-Oncology. 135 (3), 423-432 (2017).
  3. Raj, D., Agrawal, P., Gaitsch, H., Wicks, E., Tyler, B. Pharmacological strategies for improving the prognosis of glioblastoma. Expert Opinion on Pharmacotherapy. 22 (15), 2019-2031 (2021).
  4. Alomari, S., et al. Drug repurposing for Glioblastoma and current advances in drug delivery-a comprehensive review of the literature. Biomolecules. 11 (12), 1870 (2021).
  5. Serra, R., et al. Combined intracranial Acriflavine, temozolomide and radiation extends survival in a rat glioma model. European Journal of Pharmaceutics and Biopharmaceutics : Official Journal of Arbeitsgemeinschaft fur Pharmazeutische Verfahrenstechnik eV. 170, 179-186 (2022).
  6. Tang, B., Foss, K., Lichtor, T., Phillips, H., Roy, E. Resection of orthotopic murine brain glioma. Neuroimmunology and Neuroinflammation. 8 (1), 64-69 (2021).
  7. Ozawa, T., James, C. D. Establishing intracranial brain tumor xenografts with subsequent analysis of tumor growth and response to therapy using bioluminescence imaging. Journal of Visualized Experiments. (41), e1986 (2010).
  8. Poussard, A., et al. In vivo imaging systems (IVIS) detection of a neuro-invasive encephalitic virus. Journal of Visualized Experiments. (70), e4429 (2012).
  9. Lachin, J. M. Nonparametric statistical analysis. JAMA. 323 (20), 2080-2081 (2020).
  10. Louis, K. S., Siegel, A. C. Cell viability analysis using trypan blue: manual and automated methods. Methods in Molecular Biology. 740, 7-12 (2011).
  11. Spina, R., Voss, D. M., Asnaghi, L., Sloan, A., Bar, E. E. Flow cytometry-based drug screening system for the identification of small molecules that promote cellular differentiation of Glioblastoma stem cells. Journal of Visualized Experiments. (131), e56176 (2018).
  12. Rodgers, G., et al. Virtual histology of an entire mouse brain from formalin fixation to paraffin embedding. Part 2: Volumetric strain fields and local contrast changes. Journal of Neuroscience Methods. 365, 109385 (2022).
  13. Connolly, N. P., et al. Elevated fibroblast growth factor-inducible 14 expression transforms proneural-like gliomas into more aggressive and lethal brain cancer. GLIA. 69 (9), 2199-2214 (2021).
  14. Stall, B., et al. Comparison of T2 and FLAIR imaging for target delineation in high grade gliomas. Radiation Oncology. 5, 5 (2010).
  15. Das, A., et al. Establishing a standardized method for the effective intraoperative collection and biological preservation of brain tumor tissue samples using a novel tissue preservation system: a pilot study. World Neurosurgery. , (2022).
  16. Zusman, E., et al. Tissues harvested using an automated surgical approach confirm molecular heterogeneity of Glioblastoma and enhance specimen's translational research value. Frontiers in Oncology. 9, (2019).
  17. McLaughlin, N., et al. Side-cutting aspiration device for endoscopic and microscopic tumor removal. Journal of Neurological Surgery Part B. 73 (1), 11-20 (2012).

Tags

Minimally Invasive Brain Tumor Resection Rodents High Viability Tissue Collection Preclinical Research Therapeutics Standardized Resection Minimally-invasive Approach Automated Tissue Preservation System Disease State Therapy Animal Survival Sequential And Time-lapse Therapeutic Applications Biology Of The Disease Drug Discovery Novel Therapeutics Liver Tumors Head And Neck Cancers User-friendly System Published Protocol Sedation Assessment Ophthalmic Ointment Stereotactic Frame Disinfecting The Skin
Implementation of Minimally Invasive Brain Tumor Resection in Rodents for High Viability Tissue Collection
Play Video
PDF DOI DOWNLOAD MATERIALS LIST

Cite this Article

Alomari, S., Kedda, J., Malla, A.More

Alomari, S., Kedda, J., Malla, A. P., Pacis, V., Anastasiadis, P., Xu, S., McFarland, E., Sukhon, L., Gallo, B., Rincon-Torroella, J., Ben-Shalom, N., Ames, H. M., Brem, H., Woodworth, G. F., Tyler, B. Implementation of Minimally Invasive Brain Tumor Resection in Rodents for High Viability Tissue Collection. J. Vis. Exp. (183), e64048, doi:10.3791/64048 (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