• Users Online: 705
  • Print this page
  • Email this page


 
 
Table of Contents
REVIEW
Year : 2019  |  Volume : 2  |  Issue : 1  |  Page : 3-6

Importance of iatrogenic immunosuppression in the treatment of patients with high-grade glioma with immunotherapy


1 Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
2 Department of Medicine, Oncology and Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA

Date of Web Publication1-Apr-2019

Correspondence Address:
Dr. Anna F Piotrowski
Memorial Sloan Kettering Cancer Center, New York, NY
USA
Login to access the Email id

Source of Support: None, Conflict of Interest: None


DOI: 10.4103/glioma.glioma_8_19

Rights and Permissions
  Abstract 

Treatment-related lymphopenia is a poor prognostic factor for overall survival in patients with high-grade glioma and predicts suboptimal response to immune therapies. Immunotherapy is conceptually an appealing approach in adults with high-grade glioma given that effector lymphocytes are capable of penetrating the blood–brain barrier. However, 40% of these patients develop severe lymphopenia (CD4 counts <200) following concurrent radiation and temozolomide. These low lymphocyte counts are associated with inferior survival. Research suggests that this iatrogenic immunosuppression is attributed to the inadvertent radiation of circulating lymphocytes as they traverse the irradiated field. Lymphocyte subtypes are universally affected by this radiation toxicity. These findings have been reproduced in animal studies, and clinical correlations have been demonstrated in patients with various malignancies. This lymphopenia has been linked with failure to respond to immunologic interventions. Recent insights into the etiology of this radiation-induced lymphopenia have triggered a variety of novel approaches to prevent or restore immunologic function in this patient population. These include altering radiation plans, reducing the number of lymphocytes passing through the radiation field, harvesting lymphocytes before and reinfusing them after radiation, and using growth factors to restore lymphocyte counts. This manuscript reviews critical relationships between treatment-related lymphopenia and immunotherapy outcomes in patients with high-grade gliomas and novel approaches to these issues.

Keywords: High-grade glioma, immunosuppression, immunotherapy, lymphocytes, radiation-induced lymphopenia, treatment-related lymphopenia


How to cite this article:
Piotrowski AF, Grossman SA. Importance of iatrogenic immunosuppression in the treatment of patients with high-grade glioma with immunotherapy. Glioma 2019;2:3-6

How to cite this URL:
Piotrowski AF, Grossman SA. Importance of iatrogenic immunosuppression in the treatment of patients with high-grade glioma with immunotherapy. Glioma [serial online] 2019 [cited 2023 Jun 4];2:3-6. Available from: http://www.jglioma.com/text.asp?2019/2/1/3/255156


  Introduction Top


Improvements in the outcome of patients with high-grade glioma have been extremely limited over the past several decades.[1] Despite dramatic advances in imaging and surgical care, this cancer remains incurable even if it is detected early and is surgically accessible. Similarly, extensive research focused on improving the efficacy of radiation therapy has evaluated escalating radiation doses, employing different field sizes and dose fractions, using radiation sensitizers, and implanting radiation seeds within the tumor without much progress. Despite decades of trials with chemotherapeutic and biologic agents, only temozolomide has been demonstrated to improve overall survival.[2] Unfortunately, its impact on patient outcomes has been limited. In patients with glioblastoma who have a good performance status and are eligible for clinical trials, temozolomide provides an increase in overall survival of <3 months and in the 60% of patients who are O6-methylguanine-DNA methyltransferase unmethylated; it appears to provide little to no clinical benefit.[3] Increasing the dose or duration of temozolomide has not improved outcomes, and the cure rate with aggressive multimodality therapy remains close to zero.[4] Efforts to implant chemotherapy-laden biodegradable polymers into glioblastomas at the time of resection[5] or using low intensity, alternating electric fields to disrupt cell division after completing radiation therapy,[6] have had only minimal impact on survival. The results of extensive trials of targeted and antiangiogenesis agents have been very disappointing.[7],[8] As 98% of drugs approved by the U. S. Food and Drug Administration do not enter the brain in therapeutic concentrations, the efficiency of the blood–brain barrier remains a major factor limiting the efficacy of systemically administered chemotherapeutics in this difficult to treat cancer.[9]

Given the significant barriers to progress with surgery, radiation, and chemotherapy, alternative treatment approaches are needed to improve outcomes for patients with high-grade glioma. Immunotherapy aims to stimulate the systemic immune response, and lymphocytes can penetrate the blood–brain barrier, unlike most pharmaceutical agents. However, as has been documented in early clinical trials, success with immunotherapy interventions will likely not come easily. The lack of success to date may be attributed to the low mutational burden[10] and the immunologically “cold” profile of high-grade glioma,[11] the necessary use of glucocorticoids to reduce brain edema, and the lymphotoxicity that accompanies radiation, temozolomide, and steroids. This manuscript reviews critical relationships between treatment-related lymphopenia and immunotherapy outcomes in patients with high-grade gliomas and novel approaches to these issues.


  Mechanism of Treatment-Related Lymphopenia Top


The incidence, severity, and duration of Grade III-IV lymphopenia and the association between lymphopenia and survival were first described in patients with newly diagnosed high-grade glioma following radiation and temozolomide in 2011. The National Institutes of Health-funded New Approaches to Brain Tumor Therapy found that of 96 patients with high-grade glioma, 40% became severely lymphopenic (defined as CD4 count <200 cells/mm3) and remained severely depressed for at least 1 year of follow-up. Multivariate analysis showed that lymphopenia at 2 months is an independent prognostic factor for shorter survival due to tumor progression.[12] Subsequent studies by the same investigators documented that similar lymphopenia toxicity profiles followed radiation in elderly glioblastoma patients,[13] patients with head and neck,[14],[15] lung,[16] pancreatic,[17],[18] cervical,[19] esophageal,[20],[21] rectal,[22] and pediatric malignancies.[23] Interestingly, nonsmall-cell lung cancer patients who receive upfront radiation immediately become lymphopenic, whereas those who received neoadjuvant chemotherapy only become lymphopenic when the radiation is added.[24] Moreover, the association between low lymphocyte counts and overall survival is not limited to patients with high-grade glioma.[25] In retrospect, there were many reports in the 1960s and 1970s documenting that radiation to almost any site resulted in severe and sustained lymphopenia,[26],[27] but its prognosis for poor survival was not firmly established until 2011 when the high-grade glioma study was published.

Radiation of extracorporeal circulating blood has been shown to result in profound lymphopenia in multiple animal studies as well as a series of transplant patients. Dr. Cronkite et al.'s group[28] at the Brookhaven National Laboratory established that severe and long-lasting lymphopenia in calves can be produced by radiating an externalized catheter connecting an artery and a vein or the thoracic duct and a vein. Oldendorf et al.[29] described a surgical technique for delivering ionizing radiation to a large artery graft within a metal shield in living dogs. In 1965, Wolf and Hume[30] used a radioactive intra-arterial implant to produce lymphopenia in dogs. Storb et al.[31] carried out continuous extracorporeal irradiation of blood and noted lymphoid hypoplasia in addition to lymphocytopenia in baboons. These investigators reviewed hematoxylin and eosin-stained sections of lymph nodes and noted decreased cellularity and atrophic germinal centers immediately after radiation and normalization after 5–8 weeks. In the early 1970s, when potent immunosuppressive drugs were unavailable, patients awaiting kidney transplant had their peripheral blood circulating in dialysis machines irradiated to effectively induce sustained immunosuppression.[32]

A newly developed animal model of radiation-induced lymphopenia following cranial irradiation has recently been published. Using a small animal radiation research platform, C57 BL/6 and BALB/c mice were given focal brain irradiation at multiple doses and fractionation schemes: 1, 2, 3 Gy × 5 fractions, 2 Gy × 30 fractions, and 4 Gy × 10 fractions. In this study, there was no significant radiation exposure to extracranial sites such as lymphoid organs or bone marrow. Similar to humans, the mice became lymphopenic after the radiation and lost 70% of their circulating lymphocytes. Unlike humans, lymphocyte counts in these mice recovered 3 weeks following the completion of radiation. Postmortem examinations of inguinal lymph nodes, far outside of the radiation field, demonstrated remarkable histopathologic changes demonstrating systemic lymphodepletion.[33]

Recent studies strongly suggest that this iatrogenic immunosuppression occurs largely due to the inadvertent radiation of circulating lymphocytes as they pass through the radiation beam. Lymphocytes are the most radiosensitive cells in the body. A mathematical model has been published which estimates the radiation dose that lymphocytes receive while passing through the radiation field. A typical high-grade glioma treatment plan (8 cm tumor, 60 Gy in 30 fractions) was constructed using the Pinnacle radiation planning system (Philips Healthcare, Baltimore, MD, USA). Radiation doses to circulating cells were analyzed using MatLab (MathWorks, Natick, MA, USA). The model determined that radiation from the first fraction of radiation will kill 6% of all circulating lymphocytes and that with the typical 30 fractions over 98% of circulating lymphocytes had received the lymphotoxic dose.[34] A prospective immunophenotypic analysis of 20 patients with glioblastoma treated with standard radiation and temozolomide demonstrated that all subsets of B-cells, T-cells, and natural killer cells are severely decreased by standard radiation treatment.[35] The clinical, animal, and modeling studies described above strongly suggest that circulating blood should now be considered an “organ at risk” for radiation toxicities based on its effect on the immune system.[36],[37]

The immune system has long been thought to play a key role in cancer surveillance and therapy. This concept was pioneered by Dr. Coley[38] in 1890 when he injected bacteria directly into inoperable tumors in an attempt to stimulate an immune response against the cancer. This belief continues to underpin the current enthusiasm regarding the use of novel immunotherapy, such as checkpoint inhibitors.[11] If one believes that enhancing the capacity of the immune system to recognize and eradicate cancer will improve survival, then it is equally apparent that accidental injury to the immune system might compromise clinical outcomes. As outlined above, radiation to circulating lymphocytes results in a severe, persistent, and unintended injury to the immune system. The severe lymphopenia occurs in about 40% of treated patients and is associated with reduced survival which is likely due to the patient's impaired immune system to control growth of the cancer. As lymphocytes are the effector cells required for a response to immunotherapy, treatment-related lymphopenia clearly has a significant detriment on response to checkpoint inhibitor therapy. This is highlighted by a recent study of 167 patients with solid tumors who received either nivolumab or pembrolizumab. Those with persistent lymphopenia, usually after radiation therapy, had fewer responses, fewer immune side effects, and a shorter time to progression than those with higher lymphocyte counts.[39]


  Strategies to Restore Immune Function Top


Immunotherapy trials in patients with high-grade glioma face a number of significant challenges including their low mutational burden, immunologically “cold” profile of high-grade glioma, and the frequent use of glucocorticoids to control peritumoral vasogenic edema.[11] While these tumor-associated factors are unmodifiable, recent insights into the etiology of radiation-induced lymphopenia have generated optimism that this can be effectively prevented or repaired using a variety of strategies. These are briefly outlined below.

Novel radiation approaches

The mathematical model described above has identified important and actionable variables that contribute to the immunosuppression. The radiation dose delivered to circulating blood with a brain treatment plan is likely to be primarily dependent on volume of the radiation field, number of radiation fractions, and dose rate.[36] Studies in other systemic cancers have clearly shown that reducing the size of the treatment field (i.e., using stereotactic radiation) and reducing the number of administered fractions have a significant effect on posttreatment lymphocyte counts.[40],[41] An interesting experimental approach uses flash radiation, which provides radiation in milliseconds rather than minutes, and is likely to dramatically reduce radiation exposure to circulating lymphocytes.[42]

Novel prevention approaches

Two recently completed feasibility trials in patients with high-grade glioma have explored innovative ways to spare lymphocytes from radiation. The first used a sphingosine-1-phosphate inhibitor which homes lymphocytes in lymphoid organs resulting in approximately a 70% reduction in the total number of circulating lymphocytes. This study was designed to determine if the combination of this sphingosine-1-phosphate inhibitor and radiation with temozolomide resulted in opportunistic infections that would limit its utility. No severe opportunistic infections were observed clearing the way for further prospective studies (NCT02490930). A second study evaluated the feasibility of harvesting lymphocytes before radiation and reinfusing them on completion of radiation. This was also found to be safe; however, the number of lymphocytes harvested from one peripheral line was less than ideal.[43]

Novel restoration approaches

Interleukin-7 is a major regulator of lymphocytes and specifically CD4 counts. Compensatory high levels of this cytokine should be noted when CD4 counts are low and undetectable levels are seen when there are adequate numbers of these lymphocytes. This has been well documented in patients with acquired immune deficiency syndrome, sepsis, and in chemotherapy-induced lymphopenia. However, patients with high-grade glioma and severe lymphopenia from radiation have been found to have very low interleukin-7 levels for reasons that are not understood.[44] This information has led to a currently active clinical trial within the NIH-funded Adult Brain Tumor Consortium which is conducting the first study of interleukin-7 in severely lymphopenic patients when they complete 6 weeks of radiation with temozolomide (NCT03687957).


  Conclusion Top


Iatrogenic immunosuppression secondary to radiation therapy adds another challenge to the treatment of patients with high-grade glioma with immunotherapy. However, unlike those unmodifiable factors such as low mutational burden, a “cold” tumor, and the need to use glucocorticoids to treat peritumoral brain edema, there are encouraging efforts underway to prevent or repair inadvertent damage to the immune system from radiation. Priority should be given to this line of research to optimize the efficacy of immunotherapy in this aggressive cancer.

Financial support and sponsorship

This work was funded in part by the NIH/NCI Cancer Center Support Grant P30 CA008748.

Conflicts of interest

There are no conflicts of interest.



 
  References Top

1.
Aldape K, Brindle KM, Chesler L, Chopra R, Gajjar A, Gilbert MR, et al. Challenges to curing primary brain tumours. Nat Rev Clin Oncol 2019. doi: 10.1038/s41571-019-0177-5.  Back to cited text no. 1
    
2.
Stupp R, Mason WP, van den Bent MJ, Weller M, Fisher B, Taphoorn MJ, et al. Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N Engl J Med 2005;352:987-96.  Back to cited text no. 2
    
3.
Hegi ME, Diserens AC, Gorlia T, Hamou MF, de Tribolet N, Weller M, et al. MGMT gene silencing and benefit from temozolomide in glioblastoma. N Engl J Med 2005;352:997-1003.  Back to cited text no. 3
    
4.
Gilbert MR, Wang M, Aldape KD, Stupp R, Hegi ME, Jaeckle KA, et al. Dose-dense temozolomide for newly diagnosed glioblastoma: A randomized phase III clinical trial. J Clin Oncol 2013;31:4085-91.  Back to cited text no. 4
    
5.
Westphal M, Hilt DC, Bortey E, Delavault P, Olivares R, Warnke PC, et al. A phase 3 trial of local chemotherapy with biodegradable carmustine (BCNU) wafers (Gliadel wafers) in patients with primary malignant glioma. Neuro Oncol 2003;5:79-88.  Back to cited text no. 5
    
6.
Stupp R, Taillibert S, Kanner A, Read W, Steinberg D, Lhermitte B, et al. Effect of tumor-treating fields plus maintenance temozolomide vs. maintenance temozolomide alone on survival in patients with glioblastoma: A randomized clinical trial. JAMA 2017;318:2306-16.  Back to cited text no. 6
    
7.
Gilbert MR, Dignam JJ, Armstrong TS, Wefel JS, Blumenthal DT, Vogelbaum MA, et al. A randomized trial of bevacizumab for newly diagnosed glioblastoma. N Engl J Med 2014;370:699-708.  Back to cited text no. 7
    
8.
Chinot OL, Wick W, Mason W, Henriksson R, Saran F, Nishikawa R, et al. Bevacizumab plus radiotherapy-temozolomide for newly diagnosed glioblastoma. N Engl J Med 2014;370:709-22.  Back to cited text no. 8
    
9.
Pardridge WM. The blood-brain barrier: Bottleneck in brain drug development. NeuroRx 2005;2:3-14.  Back to cited text no. 9
    
10.
Brennan CW, Verhaak RG, McKenna A, Campos B, Noushmehr H, Salama SR, et al. The somatic genomic landscape of glioblastoma. Cell 2013;155:462-77.  Back to cited text no. 10
    
11.
Lim M, Xia Y, Bettegowda C, Weller M. Current state of immunotherapy for glioblastoma. Nat Rev Clin Oncol 2018;15:422-42.  Back to cited text no. 11
    
12.
Grossman SA, Ye X, Lesser G, Sloan A, Carraway H, Desideri S, et al. Immunosuppression in patients with high-grade gliomas treated with radiation and temozolomide. Clin Cancer Res 2011;17:5473-80.  Back to cited text no. 12
    
13.
Mendez JS, Govindan A, Leong J, Gao F, Huang J, Campian JL. Association between treatment-related lymphopenia and overall survival in elderly patients with newly diagnosed glioblastoma. J Neurooncol 2016;127:329-35.  Back to cited text no. 13
    
14.
Campian JL, Sarai G, Ye X, Marur S, Grossman SA. Association between severe treatment-related lymphopenia and progression-free survival in patients with newly diagnosed squamous cell head and neck cancer. Head Neck 2014;36:1747-53.  Back to cited text no. 14
    
15.
Liu LT, Chen QY, Tang LQ, Guo SS, Guo L, Mo HY, et al. The prognostic value of treatment-related lymphopenia in nasopharyngeal carcinoma patients. Cancer Res Treat 2018;50:19-29.  Back to cited text no. 15
    
16.
Tang C, Liao Z, Gomez D, Levy L, Zhuang Y, Gebremichael RA, et al. Lymphopenia association with gross tumor volume and lung V5 and its effects on non-small cell lung cancer patient outcomes. Int J Radiat Oncol Biol Phys 2014;89:1084-91.  Back to cited text no. 16
    
17.
Balmanoukian A, Ye X, Herman J, Laheru D, Grossman SA. The association between treatment-related lymphopenia and survival in newly diagnosed patients with resected adenocarcinoma of the pancreas. Cancer Invest 2012;30:571-6.  Back to cited text no. 17
    
18.
Wild AT, Ye X, Ellsworth SG, Smith JA, Narang AK, Garg T, et al. The association between chemoradiation-related lymphopenia and clinical outcomes in patients with locally advanced pancreatic adenocarcinoma. Am J Clin Oncol 2015;38:259-65.  Back to cited text no. 18
    
19.
Wu ES, Oduyebo T, Cobb LP, Cholakian D, Kong X, Fader AN, et al. Lymphopenia and its association with survival in patients with locally advanced cervical cancer. Gynecol Oncol 2016;140:76-82.  Back to cited text no. 19
    
20.
Davuluri R, Jiang W, Fang P, Xu C, Komaki R, Gomez DR, et al. Lymphocyte nadir and esophageal cancer survival outcomes after chemoradiation therapy. Int J Radiat Oncol Biol Phys 2017;99:128-35.  Back to cited text no. 20
    
21.
Fang P, Jiang W, Davuluri R, Xu C, Krishnan S, Mohan R, et al. High lymphocyte count during neoadjuvant chemoradiotherapy is associated with improved pathologic complete response in esophageal cancer. Radiother Oncol 2018;128:584-90.  Back to cited text no. 21
    
22.
Campian JL, Ye X, Sarai G, Herman J, Grossman SA. Severe treatment-related lymphopenia in patients with newly diagnosed rectal cancer. Cancer Invest 2018;36:356-61.  Back to cited text no. 22
    
23.
Miljković MD, Grossman SA, Ye X, Ellsworth S, Terezakis S. Patterns of radiation-associated lymphopenia in children with cancer. Cancer Invest 2016;34:32-8.  Back to cited text no. 23
    
24.
Campian JL, Ye X, Brock M, Grossman SA. Treatment-related lymphopenia in patients with stage III non-small-cell lung cancer. Cancer Invest 2013;31:183-8.  Back to cited text no. 24
    
25.
Grossman SA, Ellsworth S, Campian J, Wild AT, Herman JM, Laheru D, et al. Survival in patients with severe lymphopenia following treatment with radiation and chemotherapy for newly diagnosed solid tumors. J Natl Compr Canc Netw 2015;13:1225-31.  Back to cited text no. 25
    
26.
MacLennan IC, Kay HE. Analysis of treatment in childhood leukemia. IV. The critical association between dose fractionation and immunosuppression induced by cranial irradiation. Cancer 1978;41:108-11.  Back to cited text no. 26
    
27.
Raben M, Walach N, Galili U, Schlesinger M. The effect of radiation therapy on lymphocyte subpopulations in cancer patients. Cancer 1976;37:1417-21.  Back to cited text no. 27
    
28.
Cronkite EP, Jansen CR, Mather GC, Nielsen NO, Usenik EA, Adamik ER, et al. Studies on lymphocytes. I. Lymphopenia produced by prolonged extracorporeal irradiation of circulating blood. Blood 1962;20:203-13.  Back to cited text no. 28
    
29.
Oldendorf WH, Burroughs JT, Cassen B, Wetterau LW Jr. Beta radiation of circulating blood by an implanted shielded Y90 source, preliminary report of technique. J Nucl Med 1964;5:883-6.  Back to cited text no. 29
    
30.
Wolf JS, Hume DM. Studies of a method of inducing specific lymphopenia in dogs. JAMA 1965;194:1119-21.  Back to cited text no. 30
    
31.
Storb R, Ragde H, Thomas ED. Extracorporeal irradiation of the blood in baboons. Radiat Res 1969;38:43-54.  Back to cited text no. 31
    
32.
Weeke E, Thaysen JH. The effect of extracorporeal irradiation of the blood in necrokidney transplantation. Three years' follow-up study. Acta Med Scand 1974;195:485-91.  Back to cited text no. 32
    
33.
Piotrowski AF, Nirschl TR, Velarde E, Blosser L, Ganguly S, Burns KH, et al. Systemic depletion of lymphocytes following focal radiation to the brain in a murine model. Oncoimmunology 2018;7:e1445951.  Back to cited text no. 33
    
34.
Yovino S, Kleinberg L, Grossman SA, Narayanan M, Ford E. The etiology of treatment-related lymphopenia in patients with malignant gliomas: Modeling radiation dose to circulating lymphocytes explains clinical observations and suggests methods of modifying the impact of radiation on immune cells. Cancer Invest 2013;31:140-4.  Back to cited text no. 34
    
35.
Campian JL, Piotrowski AF, Ye X, Hakim FT, Rose J, Yan XY, et al. Serial changes in lymphocyte subsets in patients with newly diagnosed high grade astrocytomas treated with standard radiation and temozolomide. J Neurooncol 2017;135:343-51.  Back to cited text no. 35
    
36.
Ellsworth SG. Field size effects on the risk and severity of treatment-induced lymphopenia in patients undergoing radiation therapy for solid tumors. Adv Radiat Oncol 2018;3:512-9.  Back to cited text no. 36
    
37.
Venkatesulu BP, Mallick S, Lin SH, Krishnan S. A systematic review of the influence of radiation-induced lymphopenia on survival outcomes in solid tumors. Crit Rev Oncol Hematol 2018;123:42-51.  Back to cited text no. 37
    
38.
Coley WB. The diagnosis and treatment of bone sarcoma. Glasgow Med J 1936;126:49-86.  Back to cited text no. 38
    
39.
Diehl A, Yarchoan M, Hopkins A, Jaffee E, Grossman SA. Relationships between lymphocyte counts and treatment-related toxicities and clinical responses in patients with solid tumors treated with PD-1 checkpoint inhibitors. Oncotarget 2017;8:114268-80.  Back to cited text no. 39
    
40.
Wild AT, Herman JM, Dholakia AS, Moningi S, Lu Y, Rosati LM, et al. Lymphocyte-sparing effect of stereotactic body radiation therapy in patients with unresectable pancreatic cancer. Int J Radiat Oncol Biol Phys 2016;94:571-9.  Back to cited text no. 40
    
41.
Saito T, Toya R, Matsuyama T, Semba A, Oya N. Dosimetric predictors of treatment-related lymphopenia induced by palliative radiotherapy: Predictive ability of dose-volume parameters based on body surface contour. Radiol Oncol 2017;51:228-34.  Back to cited text no. 41
    
42.
Durante M, Bräuer-Krisch E, Hill M. Faster and safer? FLASH ultra-high dose rate in radiotherapy. Br J Radiol 2018;91:20170628.  Back to cited text no. 42
    
43.
Campian JL, Ye X, Gladstone DE, Ambady P, Nirschl TR, Borrello I, et al. Pre-radiation lymphocyte harvesting and post-radiation reinfusion in patients with newly diagnosed high grade gliomas. J Neurooncol 2015;124:307-16.  Back to cited text no. 43
    
44.
Ellsworth S, Balmanoukian A, Kos F, Nirschl CJ, Nirschl TR, Grossman SA, et al. Sustained CD4+ T cell-driven lymphopenia without a compensatory IL-7/IL-15 response among high-grade glioma patients treated with radiation and temozolomide. Oncoimmunology 2014;3:e27357.  Back to cited text no. 44
    




 

Top
 
  Search
 
    Similar in PUBMED
   Search Pubmed for
   Search in Google Scholar for
 Related articles
    Access Statistics
    Email Alert *
    Add to My List *
* Registration required (free)  

 
  In this article
Abstract
Introduction
Mechanism of Tre...
Strategies to Re...
Conclusion
References

 Article Access Statistics
    Viewed4657    
    Printed231    
    Emailed0    
    PDF Downloaded339    
    Comments [Add]    

Recommend this journal