Skip to main content
Erschienen in: Strahlentherapie und Onkologie 4/2019

Open Access 11.02.2019 | Case Study

Radiochemotherapy combined with NK cell transfer followed by second-line PD-1 inhibition in a patient with NSCLC stage IIIb inducing long-term tumor control: a case study

verfasst von: Konrad Kokowski, Stefan Stangl, Sophie Seier, Martin Hildebrandt, Peter Vaupel, Gabriele Multhoff

Erschienen in: Strahlentherapie und Onkologie | Ausgabe 4/2019

Abstract

Background

Membrane heat shock protein 70 (mHsp70) is indicative of high-risk tumors and serves as a tumor-specific target for natural killer (NK) cells stimulated with Hsp70 peptide (TKD) and Interleukin(IL)-2. Radiochemotherapy (RCT), mHsp70-targeting NK cells, and programmed death(PD)-1 inhibition were combined to improve the efficacy of tumor-specific immune cells in a non-small cell lung carcinoma (NSCLC) patient.

Patient

Following simultaneous RCT (64.8 Gy), a patient with inoperable NSCLC (cT4, cN3, cM0, stage IIIb) was treated with 4 cycles of autologous ex vivo TKD/IL-2-activated NK cells and the PD-1 antibody nivolumab as a second-line therapy. Blood samples were taken for immunophenotyping during the course of therapy.

Results

Adoptive transfer of ex vivo TKD/IL-2-activated NK cells after RCT combined with PD-1 blockade is well tolerated and results in superior overall survival (OS). No viable tumor cells but a massive immune cell infiltration in fibrotic tissue was detected after therapy. Neither tumor progression nor distant metastases were detectable by CT scanning 33 months after diagnosis. Therapy response was associated with significantly increased CD3/NKG2D+/CD94+ NK cell counts, elevated CD8+ to CD4+ T cell and CD3/CD56bright to CD3/CD56dim NK cell ratios, and significantly reduced regulatory T cells (Tregs) in the peripheral blood.

Conclusion

A combined therapy consisting of RCT, mHsp70-targeting NK cells, and PD-1 antibody inhibition is well tolerated, induces anti-tumor immunity, and results in long-term tumor control in one patient with advanced NSCLC. Further, randomized studies are necessary to confirm the efficacy of this combination therapy.

Introduction

Stress-inducible Hsp70 is frequently overexpressed in the cytosol and presented on the plasma membrane of high-risk tumors including locally advanced lung cancer and therefore serves as a universal tumor biomarker [1]. Despite combined treatment regimens consisting of radio- and (cisplatinum-based) chemotherapy (RCT), most patients with non-operable, advanced NSCLC show disease progression and poor overall survival [25]. Chronic inflammation, anti-apoptotic pathways, and nuclear factor kappa-light chain-enhancer of activated B cells(NFκB)-, hypoxia-inducible factor(HIF)-, and signal transducer and activator of transcription(STAT)- driven [6, 7] immunosuppressive mechanisms [8] can thwart anti-tumor immune responses. A major breakthrough has been the blockade of immune checkpoint inhibitors, including PD-1/PD-L1 (programnmed cell death ligand-1), providing inhibitory feedback loops for immune-mediated tumor rejection [9, 10]. In healthy individuals, checkpoint inhibitors prevent autoimmunity, whereas in cancer patients, they abrogate cytolytic and migratory activities of T and NK cells [11, 12]. Nivolumab, a fully humanized IgG4 antibody, targets PD-1 and thereby attenuates inhibitory signals [9, 11], resulting in objective tumor responses [13, 14]. In melanoma and glioblastoma cells, RCT has been found to upregulate PDL-1 expression [15]. Despite promising clinical results in NSCLC patients after PDL-1 antibody therapy [10], a relevant proportion of patients do not respond to therapy. This might be partly due to the absence of anti-tumor-specific effector cells. Therefore, anti-Hsp70-activated NK cells were combined with anti-PD-1 inhibition in a patient with advanced NSCLC after RCT.

Methods

Ethics, patient characteristics, therapies

Written informed consent was obtained from the patient and the clinical trial protocol (NSCLC-TKD/IL-2 EudraCT-No.: 2008-002130-30) was approved by the institutional ethical review board of the Klinikum rechts der Isar, TU München (TUM). A 58-year-old male smoker was diagnosed with inoperable, stage IIIb squamous NSCLC (cT4, cN3, cM0; Karnofsky >90%) in 11/2015. The patient was treated with simultaneous cisplatinum/vinorelbine-based RCT (11/2015–02/2016) with a total radiation dose of 64.8 Gy (single fractions of 1.8 Gy). Following RCT and CT scanning, the patient received 4 cycles of ex vivo TKD/IL-2-stimulated, autologous NK cells (3/2016–6/2016) on a monthly basis. Sixteen months after diagnosis (3/2017–4/2017), the patient received 3 cycles nivolumab (Bristol-Myers Squibb, Princeton, NJ, USA; 3 mg/kg body weight, total dose 200 mg), as second-line therapy. Blood samples were taken between 0 and 20 months (V0, diagnosis; V1, CT after RCT; V2, NK cell therapy, V3–V5, CT after RCT and NK cell therapy; V6, nivolumab therapy; V7, CT-guided bronchoscopy). Radiographic responses of the tumor were staged according to RECIST1.1 criteria.

Ex vivo stimulation of NK cells with TKD/IL-2

Following RCT and CT scanning, leukocyte concentrates were obtained by a 3–4 h leukapheresis (Cobe Spectra, Heimstetten, Germany) at the University Hospital Regensburg, Germany. PBLs were isolated by density gradient centrifugation in a closed SEPAX system (Biosafe, Eysins, Switzerland) and resuspended in CellGro SCGM stem cell medium (CellGenix, Freiburg/Breisgau, Germany). After counting, 5–10 × 106 PBL/ml in CellGro medium were incubated with 2 µg/ml GMP-grade TKD peptide and 100 IU/ml recombinant IL-2 (Proleukin, Novartis, Nürnberg, Germany) [16], transferred into 250 ml Teflon bags (Vue-Life-118, CellGenix, Freiburg/Breisgau, Germany), and cultured in an incubator (Heraeus, Nürnberg, Germany) under gentle rotation at 37 °C, 5% CO2, in a humidified atmosphere (90%) for 3–5 days in a GMP laboratory (TUM Cells, Munich, Germany) according to a protocol of a phase I/II clinical trial [17, 18]. Activated cells were harvested, washed twice, and resuspended in Ringer’s lactate solution (500 ml) substituted with 0.1% human serum albumin (HSA). Sterility testing of intermediate and end products was performed regularly before and after reinfusion. Between 12 and 24 h after cell preparation, cells were reinfused by iv injection within 30–60 min using a stem cell reinfusion set. Tumor staging was performed by computed tomography (CT) and/or positron-emission tomography PET/CT. Bronchoscopies were taken under CT-guidance.

Laboratory parameters and flow cytometric analysis of peripheral blood lymphocytes

Routine laboratory parameters (differential blood counts, RBC parameters, white blood cell counts), blood chemistry (creatinine, AST/SGOT, ALT/SGPT, γ‑GT, LDH) were determined after each treatment and in the follow-up period. Lymphocyte subpopulations were measured by flow cytometry on a FACSCalibur instrument (BD Biosciences, San Jose, CA, USA) in the peripheral blood of the patient at the different visits (V0–V7) with different antibody combinations (Supplementary Table 1): CD3/CD19+ B cells, CD3+ T cells, CD3+/CD4+ helper T cells, CD3+/CD8+ cytotoxic T cells, CD3+/CD56+ NKT cells, CD3+/CD4+/FoxP3+, CD3+/CD8+/FoxP3+ regulatory T cells, CD3/NKG2D+ NK cells, CD3/NKp30+ NK cells, CD3/NKp46+ NK cells, CD3/CD94+ NK cells, CD3/CD56bright; and CD3/CD56dim NK cells.
Table 1
Number of re-infused total white blood cells (WBC), total lymphocytes, total CD3/CD56+ NK cell counts, and percentage of lymphocytes and CD3/CD56+, CD3/CD56dim, and CD3/CD56bright NK cells at reinfusion cycle 1 to 4. Viability of the reinfused cells was always >92%
Cycle
1
2
3
4
WBC (× 109)
1.2
2.8
3.8
3.2
Total lymphocytes (× 109)
1.2
2.5
3.5
2.9
Total CD3/CD56+ NK cells (× 108)
1.7
3.5
5.3
4.0
CD3/CD56+ NK cells (%; normal range: 5–35%)
10
14
15
16
CD3/CD56dim NK cells (%)
2
3
2
3
CD3/CD56bright NK cells (%)
9
11
13
13

Measurement of serum Hsp70 levels

Serum (S-Monovette 7.5 ml Z, Sarstedt, Nürnbrecht, Germany) was obtained after centrifugation of peripheral blood (10 min at 4000 rpm). Aliquots (300 µl) were prepared and directly stored at −80 °C. Hsp70 serum concentrations were determined using the lipHsp70 ELISA [20] and the commercial Duo Set ELISA-kit (R&D Systems, Wiesbaden, Germany) following the manufacturer’s instructions.

Immunohistochemistry (IHC)

For IHC, formalin-fixed, paraffin-embedded (FFPE) specimens (4 µm) were stained on a Ventanas Benchmark XT using CD3- and CD56-specific antibodies (BD).

Statistics

The Student’s t-test was used since the data were normally distributed. In all experiments, differences were considered as statistically significant at a value p < 0.05.

Results

Clinical responses of a patient with NSCLC to combined NK cell and nivolumab therapy after RCT

A 58-year-old male patient was diagnosed with histologically proven, inoperable, stage IIIb squamous NSCLC (cT4, cN3, cM0; Karnofsky >90%) in 11/2015 (V0; Fig. 1). A mHsp70-positive tumor phenotype (V0) was confirmed by elevated exosomal Hsp70 (exHsp70) serum levels (11 ± 2.4 ng/ml), as determined by the lipHsp70 ELISA [20]. The radiation plan (total dose: 64.8 Gy) is shown in Fig. 2a. H&E staining of a bronchoscopy at diagnosis (V0) revealed a typical tumor morphology with atypical mitoses (Fig. 3a). Following simultaneous RCT, a partial tumor response was assessed after 3 months by CT scanning (V1, Fig. 2b). The RCT-induced decrease in tumor size was accompanied by a drop in exHsp70 concentrations (11 ± 2.4 ng/ml to 8 ± 2.3 ng/ml to 6 ± 1.1 ng/ml, V0–V2). The amount of free Hsp70, which predominantly originates from dying tumor cells (R&D ELISA), remained unaltered between V0 and V1, but gradually increased thereafter.
Four weeks after the end of RCT, the patient received 4 iv injections of ex vivo TKD/IL-2-stimulated NK cells (V2, 3/2016–6/2016) every following month. The therapy was well tolerated, as demonstrated earlier in a phase I and a running phase II clinical trial [17, 18]. Previous data demonstrated that stimulation of PBL with TKD/IL-2 selectively stimulates CD3 NK cells but not CD3+ T cells [1618]. NK cell activation was found to be superior when non-sorted PBL instead of sorted NK cells were used for stimulation [18, 21]. The number of re-infused lymphocytes and NK cells increased from 1.2 to 2.9 × 109 and 1.7 to 4.0 × 108, respectively, between the first and fourth reinfusion cycle, and CD56bright cells dominated over CD56dim NK cells (Table 1).
CT scanning after NK therapy (V3–V5) revealed no tumor progression 12 months after diagnosis (data not shown). As Hsp70 serum concentrations increased concomitantly with C‑reactive protein (CRP) between V2 and V4 (exHsp70: 22 ± 2.6 ng/ml), it was assumed that inflammation rather than tumor growth caused this increase. Sixteen months after diagnosis (V6), a higher cell density was detected by CT scanning in the previously irradiated area of the right upper lobe of the lung (Fig. 2b). Since tumor progression was assumed based on CT scanning, the patient received nivolumab as a second-line therapy in a non-metastatic setting. However, 18 months after diagnosis (V7), IHC analysis of a CT-guided bronchoscopy revealed no signs of viable tumor cells, but fibrotic tissue which is indicative of a pseudo-progress (V7, Fig. 3b). We hypothesize that the massive infiltration of CD56+ NK and CD3+ T cells detected inside the tumor (V7) which was absent at diagnosis (V0, Fig. 4) was responsible for the increased cell density determined by CT scanning at V6. In the follow-up, no tumor progression (V8, Fig. 2b) and no distant metastasis were detectable by CT scanning 33 months after diagnosis.
Apart from a transient increase in CRP (V3–V4) and after nivolumab treatment (V6–V7), routine laboratory parameters remained within normal ranges throughout the whole therapy (data not shown).

Phenotypic characterization of patient-derived effector cells

The composition of major lymphocyte subpopulations was determined between V0 and V7 in the peripheral blood. The number of B cells, which was below the normal range at diagnosis (V0), further dropped after RCT, recovered in the follow-up period after NK cell therapy (V5), and further increased above baseline levels after nivolumab treatment (V6, Fig. 5a). The patient exhibited elevated CD8+/CD4+ ratios throughout the course of therapy (Fig. 5b). The significant decrease in CD3+/CD56+ cell counts after RCT/NK cell therapy (V0–V2, p < 0.05) reached initial levels following nivolumab therapy (V6, Fig. 5c). Immunosuppressive regulatory CD4+/CD8+ Tregs (Fig. 5d) transiently increased after NK (V3) and nivolumab therapy (V6) but dropped more than 10-fold at V7. The absolute numbers of CD3 NK cell subsets such as CD3/NKG2D+, CD3/NKp30+, and CD3/NKp46+, which are assumed to be involved in the killing of mHsp70 [16], transiently increased after RCT (V1), dropped thereafter, increased to above initial levels after nivolumab therapy (V6), and remained significantly elevated compared to V0 thereafter (p < 0.05; V7; Fig. 5e and f). The number of CD3/CD94+ NK cells was raised about 10-fold after RCT and 3‑fold after nivolumab therapy compared to initial levels. The resistance of NK cells to RCT might be due to elevated intracellular glutathione levels [20]. CD3/CD56bright NK cells which were above that of CD3/CD56dim NK cells play a central role in the recognition of mHsp70 positive tumor cells ([21]; Fig. 5g). It appears that nivolumab is supportive for CD3/CD56bright NK cells because the number of this NK cell subset nearly doubled after nivolumab therapy (Fig. 5g). In contrast, the number of the CD3/CD56dim NK cell subpopulation remained nearly unaltered throughout the therapy (Fig. 5g).
A comparison of representative flow cytometric dot plots of selected major lymphocyte subpopulations at V0 and V7, such as CD3/CD19+ B cells (9% vs. 13%), CD3+/CD8+ cytotoxic lymphocytes (48% vs. 51%), CD3+/CD4+ T helper cells (29% vs. 28%), CD3/CD94+ NK cells (2% vs. 6%), and CD3/CD56+ NK cells (10% vs. 16%; Table 1). The data showed major differences in the percentage of positively stained cells and in the receptor densities as revealed by a shift of the respective plots along the x‑ and y‑axes (Fig. 6h).

Discussion

A patient with advanced NSCLC was treated with ex vivo-stimulated NK cells and nivolumab after RCT for tumor debulking [22] and for increasing damage-associated molecular patterns (DAMPs) [23, 24] including Hsp70. Membrane Hsp70 serves as a tumor target for activated NK cells [1, 1618, 21], whereas cytosolic Hsp70 impairs apoptosis [25]. Previously, exHsp70 has been shown to predict the mHsp70 status of primary tumors and viable tumor mass in tumor-bearing mice [26] and NSCLC patients [27]. Membrane Hsp70 positivity of the patient’s tumor was confirmed by elevated exHsp70 serum concentrations.
Free Hsp70 derived from dying cells is indicative of tumor response [19]. The concentration of free Hsp70 that gradually increases between V0 and V7 (0–18 months) most likely reflects tumor cell killing, whereas the drop in exHsp70 after RCT might be associated with a reduction in viable tumor mass. A transient Hsp70 increase which occurs concomitantly with that of CRP after NK and PD-1 treatment might be attributed to therapy-induced inflammation.
Historically, RCT has been considered as immunosuppressive because of large radiation fields that included substantial volumes of the blood and bone marrow [28]. However, due to advances in radiation planning and equipment, normal tissue damage can be minimized. In preclinical models, RCT has been shown to induce immunogenic tumor cell death that might elicit abscopal effects [24, 29]. A recent study on lung cancer [11] and squamous carcinoma of the head and neck [30] demonstrated that anti-PD-1 therapies are most efficient in tumors with a high mutational load. The smoking history of the patient and the RCT-induced release of DAMPs might partly explain the beneficial outcome of the patient.
Stage III NSCLC patients after RCT in the control arm of the PACIFIC trial showed tumor progression already after 5.6 months [10]. The median OS of NSCLC patients in stage IIIA/B ranged between 28.7 and 20.3 months following standard versus high-dose conformal RCT [31, 32]. As a comparison, the NSCLC patient who received RCT, NK cells, and PD-1 was progression free for more than 35 months.
Immunophenotyping of peripheral blood lymphocytes of tumor patients by multicolor flow cytometry has been found to be useful for predicting outcome and detecting immunomodulation by ionizing irradiation [33, 34]. Similar to studies with mammary and prostate carcinomas [35], RCT also negatively impacts on the B cell compartment of the NSCLC patient, due to the sensitivity of B cells towards radiotherapy [36]. B cells, which are important players in the cross-talk of the innate and adaptive immunity [37], recovered at V5 and increased during nivolumab treatment, concomitant with the number of CD3+/CD8+, CD3+/CD4+, and CD3+/CD56+ T cells that might also contribute to protective anti-tumor immunity.
Regulatory CD4+/CD25+/FoxP3+ T cells (Tregs) are well known to attenuate T and NK cell activity by secretion of anti-inflammatory suppressive cytokines that impede cytotoxicity and migratory capacity of effector cells. Generally, tumor patients have elevated Treg counts in the peripheral blood and tumor microenvironment [38], which is associated with adverse clinical outcome. In the NSCLC patient, RCT did not immediately influence Tregs and during NK cell therapy, a transient increase of this population was observed. This finding is most likely due to an RCT-induced inflammatory response which might induce the production of IL-2. However, after nivolumab therapy (V7), Tregs dropped to below initial levels. As shown in preclinical models, a depletion of Tregs could restore T and NK cell mediated anti-tumor immunity [39].
NK cells play a crucial role in first-line defense against cancer [4043]. Apart from CD8+ cytotoxic T cells [44], studies have indicated that the OS of patients with oropharyngeal cancer positively correlates with high numbers of tumor-infiltrating CD3/CD56+ NK cells [45] that recognize antigens via activatory receptors belonging to the C‑type lectin family [21, 46, 47]. The success of an immune checkpoint inhibitor blockade is dependent on anti-tumor-specific effector cells [48, 49]. Similar to a patient with colon cancer [21, 45, 50], the stimulation with TKD/IL-2 induced a shift towards the CD3/CD56bright NK cell subset which was further propagated by nivolumab.
Apart from a complex network of inhibitory/activatory receptors with immune tyrosine-based inhibitory/activatory motives (ITIM/ITAM) [51], the presence or absence of PD-1 receptors also determines the cytolytic function of activated NK cells [52]. Therefore, inhibition of the PD-1 pathway after adoptive transfer of ex vivo-stimulated NK cells might synergistically enhance their survival and cytolytic activity. Exosomal Hsp70, which has been shown to attract NK cells in vitro [53], might further stimulate their migratory and cytolytic activity against mHsp70-positive tumors.

Acknowledgements

We thank Anett Lange for editorial support. The work was supported by BMBF “Innovative therapies” (01GU0823), BMBF “Kompetenzverbund Strahlenforschung” (02NUK038A), and BMWi (AiF project, ZF4320102CS7), German Research Foundation DFG (SFB824/3), DFG (STA1520/1-1), and the Technische Universität München (TUM) within the DFG funding program Open Access Publishing.

Conflict of interest

K. Kokowski, S. Stangl, S. Seier, M. Hildebrandt, P. Vaupel, and G. Multhoff declare that they have no competing interests.
Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

© Springer Medizin

Bis 11. April 2024 bestellen und im ersten Jahr 50 % sparen!

e.Med Radiologie

Kombi-Abonnement

Mit e.Med Radiologie erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Radiologie, den Premium-Inhalten der radiologischen Fachzeitschriften, inklusive einer gedruckten Radiologie-Zeitschrift Ihrer Wahl.

© Springer Medizin

Bis 11. April 2024 bestellen und im ersten Jahr 50 % sparen!

Strahlentherapie und Onkologie

Print-Titel

•Übersichten, Originalien, Kasuistiken

•Kommentierte Literatur aus der Radioonkologie, Strahlenbiologie und -physik

Literatur
1.
Zurück zum Zitat Stangl S, Gehrmann M, Riegger J et al (2011) Targeting membrane heat-shock protein 70 (Hsp70) on tumors by cmHsp70.1 antibody. Proc Natl Acad Sci USA 108:733–738CrossRefPubMed Stangl S, Gehrmann M, Riegger J et al (2011) Targeting membrane heat-shock protein 70 (Hsp70) on tumors by cmHsp70.1 antibody. Proc Natl Acad Sci USA 108:733–738CrossRefPubMed
2.
Zurück zum Zitat Ferlay J, Shin HR, Bray F et al (2010) Estimates of worldwide burden of cancer in 2008: GLOBOCAN 2008. Int J Cancer 127:2893–2917CrossRefPubMed Ferlay J, Shin HR, Bray F et al (2010) Estimates of worldwide burden of cancer in 2008: GLOBOCAN 2008. Int J Cancer 127:2893–2917CrossRefPubMed
3.
Zurück zum Zitat Oberije C, De Ruysscher D, Houben R et al (2015) A validated prediction model for overall survival from stage III non-small cell lung cancer: toward survival prediction for individual patients. Int J Radiat Oncol Biol Phys 92:935–944CrossRefPubMedPubMedCentral Oberije C, De Ruysscher D, Houben R et al (2015) A validated prediction model for overall survival from stage III non-small cell lung cancer: toward survival prediction for individual patients. Int J Radiat Oncol Biol Phys 92:935–944CrossRefPubMedPubMedCentral
4.
Zurück zum Zitat Ahn JS, Ahn YC, Kim JH et al (2015) Multinational randomized phase III trial with or without consolidation chemotherapy using docetaxel and cisplatin after concurrent chemoradiation in inoperable stage III NSCLC: KCSG-LU05-04. J Clin Oncol 33:2660–2666CrossRefPubMed Ahn JS, Ahn YC, Kim JH et al (2015) Multinational randomized phase III trial with or without consolidation chemotherapy using docetaxel and cisplatin after concurrent chemoradiation in inoperable stage III NSCLC: KCSG-LU05-04. J Clin Oncol 33:2660–2666CrossRefPubMed
5.
Zurück zum Zitat Huber RM, Flentje M, Schmidt M et al (2006) Simultaneous chemoradiotherapy compared with radioptherapy alone after induction chemotherapy in inoperable stage IIIA or stage IIIB NSCLC: study CTRT99/97 by the bronchial carcinoma therapy group. J Clin Oncol 24:4397–4404CrossRefPubMed Huber RM, Flentje M, Schmidt M et al (2006) Simultaneous chemoradiotherapy compared with radioptherapy alone after induction chemotherapy in inoperable stage IIIA or stage IIIB NSCLC: study CTRT99/97 by the bronchial carcinoma therapy group. J Clin Oncol 24:4397–4404CrossRefPubMed
6.
Zurück zum Zitat Vaupel P, Multhoff G (2016) Adenosine can thwart antitumor immune responses elicited by radiotherapy: therapeutic strategies alleviating protumor ADO activities. Strahlenther Onkol 192:279–287CrossRefPubMed Vaupel P, Multhoff G (2016) Adenosine can thwart antitumor immune responses elicited by radiotherapy: therapeutic strategies alleviating protumor ADO activities. Strahlenther Onkol 192:279–287CrossRefPubMed
8.
Zurück zum Zitat Schreiber RD, Old LJ, Smyth MJ (2011) Cancer immunoediting: integrating immunity’s roles in cancer suppression and promotion. Science 331:1565–1570CrossRefPubMed Schreiber RD, Old LJ, Smyth MJ (2011) Cancer immunoediting: integrating immunity’s roles in cancer suppression and promotion. Science 331:1565–1570CrossRefPubMed
10.
Zurück zum Zitat Antonia SJ, Villegas A, Daniel D et al (2017) Durvalumab after chemoradiotherapy in stage III NSCLC. New Engl J Med 377:1919–1929CrossRefPubMed Antonia SJ, Villegas A, Daniel D et al (2017) Durvalumab after chemoradiotherapy in stage III NSCLC. New Engl J Med 377:1919–1929CrossRefPubMed
11.
Zurück zum Zitat Rizvi NA, Hellmann MD, Snyder A et al (2015) Cancer immunology: Mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer. Science 348:124–128CrossRefPubMedPubMedCentral Rizvi NA, Hellmann MD, Snyder A et al (2015) Cancer immunology: Mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer. Science 348:124–128CrossRefPubMedPubMedCentral
13.
Zurück zum Zitat Brahmer JR, Drake CG, Wollner I et al (2010) Phase I study of single-agent anti-programmed death-1 (MDX-1106) in refractory solid tumors: safety, clinical activity, pharmacodynamics, and immunologic correlates. J Clin Oncol 28:3167–3175CrossRefPubMedPubMedCentral Brahmer JR, Drake CG, Wollner I et al (2010) Phase I study of single-agent anti-programmed death-1 (MDX-1106) in refractory solid tumors: safety, clinical activity, pharmacodynamics, and immunologic correlates. J Clin Oncol 28:3167–3175CrossRefPubMedPubMedCentral
14.
Zurück zum Zitat Rizvi NA, Mazieres J, Planchard D et al (2015) Activity and safety of nivolumab, an anti-PD-1 immune checkpoint inhibitor, for patients with advanced, refractory squamous non-small-cell lung cancer (checkmate 063): a phase II, single-arm trial. Lancet Oncol 16:257–265CrossRefPubMedPubMedCentral Rizvi NA, Mazieres J, Planchard D et al (2015) Activity and safety of nivolumab, an anti-PD-1 immune checkpoint inhibitor, for patients with advanced, refractory squamous non-small-cell lung cancer (checkmate 063): a phase II, single-arm trial. Lancet Oncol 16:257–265CrossRefPubMedPubMedCentral
15.
Zurück zum Zitat Derer A, Spiljar M, Bäumler M et al (2016) Chemoradiation increases PD-L1 expression in certain melanoma and glioblastoma cells. Front Immunol 7:10CrossRef Derer A, Spiljar M, Bäumler M et al (2016) Chemoradiation increases PD-L1 expression in certain melanoma and glioblastoma cells. Front Immunol 7:10CrossRef
16.
Zurück zum Zitat Multhoff G, Pfister K, Gehrmann M et al (2001) A 14-mer Hsp70 peptide stimulates natural killer (NK) cell activity. Cell Stress Chaperones 6:337–344CrossRefPubMedPubMedCentral Multhoff G, Pfister K, Gehrmann M et al (2001) A 14-mer Hsp70 peptide stimulates natural killer (NK) cell activity. Cell Stress Chaperones 6:337–344CrossRefPubMedPubMedCentral
17.
Zurück zum Zitat Krause SW, Gastpar R, Andreesen R et al (2004) Treatment of colon and lung cancer patients with ex vivo heat shock protein 70-peptide-activated, autologous natural killer cells: A clinical phase I trial. Clin Cancer Res 10:3699–3707CrossRefPubMed Krause SW, Gastpar R, Andreesen R et al (2004) Treatment of colon and lung cancer patients with ex vivo heat shock protein 70-peptide-activated, autologous natural killer cells: A clinical phase I trial. Clin Cancer Res 10:3699–3707CrossRefPubMed
18.
Zurück zum Zitat Specht HM, Ahrens N, Blankenstein C et al (2015) Heat shock protein 70 (Hsp70) peptide activated natural killer (NK) cells for the treatment of patients with non-small cell lung cancer (NSCLC) after radiochemotherapy (RCTx)—from preclinical studies to a clinical phase II trial. Front Immunol 6:162CrossRefPubMedPubMedCentral Specht HM, Ahrens N, Blankenstein C et al (2015) Heat shock protein 70 (Hsp70) peptide activated natural killer (NK) cells for the treatment of patients with non-small cell lung cancer (NSCLC) after radiochemotherapy (RCTx)—from preclinical studies to a clinical phase II trial. Front Immunol 6:162CrossRefPubMedPubMedCentral
19.
Zurück zum Zitat Breuninger S, Erl J, Knape C et al (2014) Quantitative analysis of liposomal heat shock protein 70 (Hsp70) in the blood of tumor patients using an novel LipHsp70 ELISA. J Clin Cell Immunol 5:2–10CrossRef Breuninger S, Erl J, Knape C et al (2014) Quantitative analysis of liposomal heat shock protein 70 (Hsp70) in the blood of tumor patients using an novel LipHsp70 ELISA. J Clin Cell Immunol 5:2–10CrossRef
20.
Zurück zum Zitat Multhoff G, Meier T, Botzler C et al (1995) Differential effects of ifosfamide on the capacity of cytotoxic T lymphocytes and natural killer cells to lyse their target cells correlate with intracellular glutathione levels. Blood 85:2124–2131PubMed Multhoff G, Meier T, Botzler C et al (1995) Differential effects of ifosfamide on the capacity of cytotoxic T lymphocytes and natural killer cells to lyse their target cells correlate with intracellular glutathione levels. Blood 85:2124–2131PubMed
21.
Zurück zum Zitat Gross C, Schmidt-Wolf IG, Nagaraj S et al (2003) Heat shock protein 70-reactivity is associated with increased cell surface density of CD94/CD56 on primary natural killer cells. Cell Stress Chaperones 8:348–360CrossRefPubMedPubMedCentral Gross C, Schmidt-Wolf IG, Nagaraj S et al (2003) Heat shock protein 70-reactivity is associated with increased cell surface density of CD94/CD56 on primary natural killer cells. Cell Stress Chaperones 8:348–360CrossRefPubMedPubMedCentral
22.
Zurück zum Zitat Dutz A, Troost EGC, Löck S (2018) Proton therapy not superior to IMRT in locally advanced NSCLC. Strahlenther Onkol 194:790–793CrossRefPubMed Dutz A, Troost EGC, Löck S (2018) Proton therapy not superior to IMRT in locally advanced NSCLC. Strahlenther Onkol 194:790–793CrossRefPubMed
23.
Zurück zum Zitat Martin D, Rödel C, Fokas E (2018) Nivolumab for pretreated metastatic anal cancer: immune checkpoint blockade is also advised in combination with RCT. Strahlenther Onkol 194:356–357CrossRefPubMed Martin D, Rödel C, Fokas E (2018) Nivolumab for pretreated metastatic anal cancer: immune checkpoint blockade is also advised in combination with RCT. Strahlenther Onkol 194:356–357CrossRefPubMed
24.
Zurück zum Zitat Rückert M, Deloch L, Fietkau R et al (2018) Immune modulatory effects of radiotherapy as basis for well-reasoned radioimmunotherapies. Strahlenther Onkol 194(6):509–519CrossRefPubMed Rückert M, Deloch L, Fietkau R et al (2018) Immune modulatory effects of radiotherapy as basis for well-reasoned radioimmunotherapies. Strahlenther Onkol 194(6):509–519CrossRefPubMed
25.
26.
Zurück zum Zitat Bayer C, Liebhardt ME, Schmid TE et al (2014) Validation of heat shock protein 70 as a tumor-specific biomarker for monitoring the outcome of radiation therapy in tumor mouse models. Int J Radiat Oncol Biol Phys 88:694–700CrossRefPubMed Bayer C, Liebhardt ME, Schmid TE et al (2014) Validation of heat shock protein 70 as a tumor-specific biomarker for monitoring the outcome of radiation therapy in tumor mouse models. Int J Radiat Oncol Biol Phys 88:694–700CrossRefPubMed
27.
Zurück zum Zitat Gunther S, Ostheimer C, Stangl S et al (2015) Correlation of Hsp70 serum levels with gross tumor volume and composition of lymphocyte subpopulations in patients with squamous cell and adeno non-small cell lung cancer. Front Immunol 6:e556CrossRef Gunther S, Ostheimer C, Stangl S et al (2015) Correlation of Hsp70 serum levels with gross tumor volume and composition of lymphocyte subpopulations in patients with squamous cell and adeno non-small cell lung cancer. Front Immunol 6:e556CrossRef
28.
Zurück zum Zitat Heidecke CD, Weighardt H, Feith M et al (2002) Neoadjuvant treatment of esophageal cancer: Immunosuppression following combined radiochemotherapy. Surgery 132:495–501CrossRefPubMed Heidecke CD, Weighardt H, Feith M et al (2002) Neoadjuvant treatment of esophageal cancer: Immunosuppression following combined radiochemotherapy. Surgery 132:495–501CrossRefPubMed
29.
Zurück zum Zitat Demaria S, Golden EB, Formenti SC (2015) Role of local radiation therapy in cancer immunotherapy. JAMA Oncol 1:1325–1332CrossRefPubMed Demaria S, Golden EB, Formenti SC (2015) Role of local radiation therapy in cancer immunotherapy. JAMA Oncol 1:1325–1332CrossRefPubMed
30.
Zurück zum Zitat Mandal R, Senbabaoglu Y, Desrichard A et al (2016) The head and neck cancer immune landscape and its immunotherapeutic implications. JCI Insight 1:e89829CrossRefPubMedPubMedCentral Mandal R, Senbabaoglu Y, Desrichard A et al (2016) The head and neck cancer immune landscape and its immunotherapeutic implications. JCI Insight 1:e89829CrossRefPubMedPubMedCentral
31.
Zurück zum Zitat Bradley JD, Paulus R, Komaki R et al (2015) Standard-dose versus high-dose conformal radiotherapy with concurrent and consolidation carboplatin plus paclitaxel with or without cetuximab for patients with stage IIIA or IIIB NSCLC (RTOG0617): a randomized, two-by-two factorial phase III study. Lancet Oncol 16:187–199CrossRefPubMedPubMedCentral Bradley JD, Paulus R, Komaki R et al (2015) Standard-dose versus high-dose conformal radiotherapy with concurrent and consolidation carboplatin plus paclitaxel with or without cetuximab for patients with stage IIIA or IIIB NSCLC (RTOG0617): a randomized, two-by-two factorial phase III study. Lancet Oncol 16:187–199CrossRefPubMedPubMedCentral
32.
Zurück zum Zitat Frey B, Rückert M, Deloch L et al (2017) Immunomodulation by ionizing radiation—impact for design of radio-immunotherapies and for treatment of inflammatory diseases. Immunol Rev 280:231–248CrossRefPubMed Frey B, Rückert M, Deloch L et al (2017) Immunomodulation by ionizing radiation—impact for design of radio-immunotherapies and for treatment of inflammatory diseases. Immunol Rev 280:231–248CrossRefPubMed
34.
Zurück zum Zitat Wada K, Kishi N, Kanayama N et al (2018) Radiation dose escalation in accelerated hypofractionated radiotherapy for stage III NSCLC. Anticancer Res 38:5951–5958CrossRefPubMed Wada K, Kishi N, Kanayama N et al (2018) Radiation dose escalation in accelerated hypofractionated radiotherapy for stage III NSCLC. Anticancer Res 38:5951–5958CrossRefPubMed
35.
Zurück zum Zitat Sage EK, Schmid TE, Sedelmayr M et al (2015) Comparative analysis of the effects of radiotherapy versus radiotherapy after adjuvant chemotherapy on the composition of lymphocyte subpopulations in breast cancer patients. Strahlenther Onkol 118:176–180 Sage EK, Schmid TE, Sedelmayr M et al (2015) Comparative analysis of the effects of radiotherapy versus radiotherapy after adjuvant chemotherapy on the composition of lymphocyte subpopulations in breast cancer patients. Strahlenther Onkol 118:176–180
36.
Zurück zum Zitat Belka C, Ottinger H, Kreuzfelder E et al (1999) Impact of localized radiotherapy on blood immune cells counts and function in humans. Radiother Oncol 50:199–204CrossRefPubMed Belka C, Ottinger H, Kreuzfelder E et al (1999) Impact of localized radiotherapy on blood immune cells counts and function in humans. Radiother Oncol 50:199–204CrossRefPubMed
37.
Zurück zum Zitat van Beek JJ, Gorris MA, Skold AE et al (2016) Human blood myeloid and plasmacytoid dendritic cells cross activate each other and synergize in inducing NK cell cytotoxicity. Oncoimmunology 5:e1227902CrossRefPubMedPubMedCentral van Beek JJ, Gorris MA, Skold AE et al (2016) Human blood myeloid and plasmacytoid dendritic cells cross activate each other and synergize in inducing NK cell cytotoxicity. Oncoimmunology 5:e1227902CrossRefPubMedPubMedCentral
38.
Zurück zum Zitat Fecci PE, Mitchell DA, Whitesides JF et al (2006) Increased regulatory T cell fraction amidst a diminished CD4 compartment explains cellular immune defects in patients with malignant glioma. Cancer Res 66:3294–3302CrossRefPubMed Fecci PE, Mitchell DA, Whitesides JF et al (2006) Increased regulatory T cell fraction amidst a diminished CD4 compartment explains cellular immune defects in patients with malignant glioma. Cancer Res 66:3294–3302CrossRefPubMed
39.
Zurück zum Zitat Teng MW, Ngiow SF, von Scheidt B et al (2010) Conditional regulatory T‑cell depletion releases adaptive immunity preventing carcinogenesis and suppressing established tumor growth. Cancer Res 70:7800–7809CrossRefPubMed Teng MW, Ngiow SF, von Scheidt B et al (2010) Conditional regulatory T‑cell depletion releases adaptive immunity preventing carcinogenesis and suppressing established tumor growth. Cancer Res 70:7800–7809CrossRefPubMed
41.
Zurück zum Zitat Morvan MG, Lanier LL (2016) NK cells and cancer: you can teach innate cells new tricks. Nat Rev Cancer 16:7–19CrossRefPubMed Morvan MG, Lanier LL (2016) NK cells and cancer: you can teach innate cells new tricks. Nat Rev Cancer 16:7–19CrossRefPubMed
43.
Zurück zum Zitat Pross HF, Lotzova E (1993) Role of natural killer cells in cancer. Nat Immun 12:279–292PubMed Pross HF, Lotzova E (1993) Role of natural killer cells in cancer. Nat Immun 12:279–292PubMed
44.
Zurück zum Zitat Balermpas P, Rödel F, Krause M et al (2017) The PD-1/PD-L1 axis and human papilloma virus in patients with head and neck cancer after adjuvant chemoradiotherapy: A multicentre study of the German Cancer Consortium Radiation Oncology Group (DKTK-ROG). Int J Cancer 141:594–603CrossRefPubMed Balermpas P, Rödel F, Krause M et al (2017) The PD-1/PD-L1 axis and human papilloma virus in patients with head and neck cancer after adjuvant chemoradiotherapy: A multicentre study of the German Cancer Consortium Radiation Oncology Group (DKTK-ROG). Int J Cancer 141:594–603CrossRefPubMed
45.
Zurück zum Zitat Wagner S, Wittekindt C, Reuschenbach M et al (2016) CD56-positive lymphocyte infiltration in relation to human papillomavirus association and prognostic significance in oropharyngeal squamous cell carcinoma. Int J Cancer 138:2263–2273CrossRefPubMed Wagner S, Wittekindt C, Reuschenbach M et al (2016) CD56-positive lymphocyte infiltration in relation to human papillomavirus association and prognostic significance in oropharyngeal squamous cell carcinoma. Int J Cancer 138:2263–2273CrossRefPubMed
46.
Zurück zum Zitat Kruse PH, Matta J, Ugolini S et al (2014) Natural cytotoxicity receptors and their ligands. Immunol Cell Biol 92:221–229CrossRefPubMed Kruse PH, Matta J, Ugolini S et al (2014) Natural cytotoxicity receptors and their ligands. Immunol Cell Biol 92:221–229CrossRefPubMed
47.
Zurück zum Zitat Gross C, Hansch D, Gastpar R et al (2003) Interaction of heat shock protein 70 peptide with NK cells involves the NK receptor CD94. Biol Chem 384:267–279CrossRefPubMed Gross C, Hansch D, Gastpar R et al (2003) Interaction of heat shock protein 70 peptide with NK cells involves the NK receptor CD94. Biol Chem 384:267–279CrossRefPubMed
48.
Zurück zum Zitat Ariffin AB, Forde PF, Jahangeer S et al (2014) Releasing pressure in tumors: what do we know so far and where do we go from here? A review. Cancer Res 74:2655–2662CrossRefPubMed Ariffin AB, Forde PF, Jahangeer S et al (2014) Releasing pressure in tumors: what do we know so far and where do we go from here? A review. Cancer Res 74:2655–2662CrossRefPubMed
49.
Zurück zum Zitat Zitvogel L, Tesniere A, Kroemer G (2006) Cancer despite immunosurveillance: immunoselection and immunosubversion. Nat Rev Immunol 6:715–727CrossRefPubMed Zitvogel L, Tesniere A, Kroemer G (2006) Cancer despite immunosurveillance: immunoselection and immunosubversion. Nat Rev Immunol 6:715–727CrossRefPubMed
50.
Zurück zum Zitat Milani V, Stangl S, Issels R et al (2009) Anti-tumor activity of patient derived NK cells after cell-based immunotherapy—a case report. J Transl Med 7:50CrossRefPubMedPubMedCentral Milani V, Stangl S, Issels R et al (2009) Anti-tumor activity of patient derived NK cells after cell-based immunotherapy—a case report. J Transl Med 7:50CrossRefPubMedPubMedCentral
51.
Zurück zum Zitat Martinet L, Smyth MJ (2015) Balancing natural killer cell activation through paired receptors. Nat Rev Immunol 15:243–254CrossRefPubMed Martinet L, Smyth MJ (2015) Balancing natural killer cell activation through paired receptors. Nat Rev Immunol 15:243–254CrossRefPubMed
52.
Zurück zum Zitat Benson DM Jr., Bakan CE, Mishra A et al (2010) The PD-1/PD-L1 axis modulates the natural killer cell versus multiple myeloma effect: a therapeutic target for CT-011, a novel monoclonal anti-PD-1 antibody. Blood 116:2286–2294CrossRefPubMedPubMedCentral Benson DM Jr., Bakan CE, Mishra A et al (2010) The PD-1/PD-L1 axis modulates the natural killer cell versus multiple myeloma effect: a therapeutic target for CT-011, a novel monoclonal anti-PD-1 antibody. Blood 116:2286–2294CrossRefPubMedPubMedCentral
53.
Zurück zum Zitat Gastpar R, Gehrmann M, Bausero MA et al (2005) Heat shock protein 70 surface-positive tumor exosomes stimulate migratory and cytolytic activity of NK cells. Cancer Res 65:5238–5247CrossRefPubMedPubMedCentral Gastpar R, Gehrmann M, Bausero MA et al (2005) Heat shock protein 70 surface-positive tumor exosomes stimulate migratory and cytolytic activity of NK cells. Cancer Res 65:5238–5247CrossRefPubMedPubMedCentral
Metadaten
Titel
Radiochemotherapy combined with NK cell transfer followed by second-line PD-1 inhibition in a patient with NSCLC stage IIIb inducing long-term tumor control: a case study
verfasst von
Konrad Kokowski
Stefan Stangl
Sophie Seier
Martin Hildebrandt
Peter Vaupel
Gabriele Multhoff
Publikationsdatum
11.02.2019
Verlag
Springer Berlin Heidelberg
Erschienen in
Strahlentherapie und Onkologie / Ausgabe 4/2019
Print ISSN: 0179-7158
Elektronische ISSN: 1439-099X
DOI
https://doi.org/10.1007/s00066-019-01434-9

Weitere Artikel der Ausgabe 4/2019

Strahlentherapie und Onkologie 4/2019 Zur Ausgabe

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.